1
|
Cao M, Zou J, Shi M, Zhao D, Liu C, Liu Y, Li L, Jiang H. A promising therapeutic: Exosome-mediated mitochondrial transplantation. Int Immunopharmacol 2024; 142:113104. [PMID: 39270344 DOI: 10.1016/j.intimp.2024.113104] [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: 07/17/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024]
Abstract
Mitochondrial dysfunction has been identified as a trigger for cellular autophagy dysfunction and programmed cell death. Emerging studies have revealed that, in pathological contexts, intercellular transfer of mitochondria takes place, facilitating the restoration of mitochondrial function, energy metabolism, and immune homeostasis. Extracellular vesicles, membranous structures released by cells, exhibit reduced immunogenicity and enhanced stability during the transfer of mitochondria. Thus, this review provides a concise overview of mitochondrial dysfunction related diseases and the mechanism of mitochondrial dysfunction in diseases progression, and the composition and functions of the extracellular vesicles, along with elucidating the principal mechanisms underlying intercellular mitochondrial transfer. In this article, we will focus on the advancements in both animal models and clinical trials concerning the therapeutic efficacy of extracellular vesicle-mediated mitochondrial transplantation across various systemic diseases in neurodegenerative diseases and cardiovascular diseases. Additionally, the review delves into the multifaceted roles of extracellular vesicle-transplanted mitochondria, encompassing anti-inflammatory actions, promotion of tissue repair, enhancement of cellular function, and modulation of metabolic and immune homeostasis within diverse pathological contexts, aiming to provide novel perspectives for extracellular vesicle transplantation of mitochondria in the treatment of various diseases.
Collapse
Affiliation(s)
- Meiling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiahui Zou
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Mingyue Shi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Danyang Zhao
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Chang Liu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yanshan Liu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
2
|
Liu Y, Min Z, Mo J, Ju Z, Chen J, Liang W, Zhang L, Li H, Chan GCF, Wei Y, Zhang W. ExomiRHub: A comprehensive database for hosting and analyzing human disease-related extracellular microRNA transcriptomics data. Comput Struct Biotechnol J 2024; 23:3104-3116. [PMID: 39219717 PMCID: PMC11362623 DOI: 10.1016/j.csbj.2024.07.024] [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: 04/15/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular microRNA (miRNA) expression data generated by different laboratories exhibit heterogeneity, which poses challenges for biologists without bioinformatics expertise. To address this, we introduce ExomiRHub (http://www.biomedical-web.com/exomirhub/), a user-friendly database designed for biologists. This database incorporates 191 human extracellular miRNA expression datasets associated with 112 disease phenotypes, 62 treatments, and 24 genotypes, encompassing 29,198 and 23 sample types. ExomiRHub also integrates 16,012 miRNA transcriptomes of 156 cancer subtypes from The Cancer Genome Atlas. All the data in ExomiRHub were further standardized and curated with annotations. The platform offers 25 analytical functions, including differential expression, co-expression, Weighted Gene Co-Expression Network Analysis (WGCNA), feature selection, and functional enrichment, enabling users to select samples, define groups, and customize parameters for analyses. Moreover, ExomiRHub provides a web service that allows biologists to analyze their uploaded miRNA expression data. Four additional tools were developed to evaluate the functions and targets of miRNAs and miRNA variations. Through ExomiRHub, we identified extracellular miRNA biomarkers associated with angiogenesis for monitoring glioma progression, demonstrating its potential to significantly accelerate the discovery of extracellular miRNA biomarkers.
Collapse
Affiliation(s)
- Yang Liu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou 510182, China
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Bioinformatics, Outstanding Biotechnology Co., Ltd.-Shenzhen, Shenzhen 518026, China
| | - Zhuochao Min
- School of Zoology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- School of Information and Software Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jing Mo
- Department of Bioinformatics, Outstanding Biotechnology Co., Ltd.-Shenzhen, Shenzhen 518026, China
| | - Zhen Ju
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jianliang Chen
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Weiling Liang
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Lantian Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou 510182, China
| | - Hanguang Li
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Godfrey Chi-Fung Chan
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yanjie Wei
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenliang Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou 510182, China
- Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Pediatrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Bioinformatics, Outstanding Biotechnology Co., Ltd.-Shenzhen, Shenzhen 518026, China
| |
Collapse
|
3
|
Wang JL, Huang QM, Hu DX, Zhang WJ. Therapeutic effect of exosomes derived from Schwann cells in the repair of peripheral nerve injury. Life Sci 2024; 357:123086. [PMID: 39357794 DOI: 10.1016/j.lfs.2024.123086] [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: 07/18/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Peripheral nerve injury (PNI) can cause nerve demyelination, neuronal apoptosis, axonal atrophy, inflammatory infiltration, glial scar formation, and other pathologies that can lead to sensory and motor dysfunction and seriously affect the psychosomatic health of patients. There is currently no effective treatment method, so exploring a promising treatment method is of great significance. Several studies have revealed the therapeutic roles of Schwann cells (SCs) and their exosomes in nerve injury repair. Exosomes are extracellular nanovesicles secreted by cells that act as key molecules in intercellular communication. Progress has been made in understanding the role of exosomes derived from SCs (SC-EXOs) in peripheral nerve regeneration, including the promotion of axonal regeneration and myelin formation, anti-inflammation, vascular regeneration, neuroprotection, and neuroregulation. Therefore, in this paper, we summarize the functional characteristics of SC-EXOs and discuss their potential therapeutic effects on PNI repair as well as some existing problems and future challenges.
Collapse
Affiliation(s)
- Jia-Ling Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Qi-Ming Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
4
|
Baran Z, Çetinkaya M, Baran Y. Mesenchymal Stem Cells in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39470980 DOI: 10.1007/5584_2024_824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
The mesenchymal stem/stromal cells (MSCs) are multipotent cells that were initially discovered in the bone marrow in the late 1960s but have so far been discovered in almost all tissues of the body. The multipotent property of MSCs enables them to differentiate into various cell types and lineages, such as adipocytes, chondrocytes, and osteocytes. The immunomodulation capacity and tumor-targeting features of MSCs made their use crucial for cell-based therapies in cancer treatment, yet limited advancement could be observed in translational medicine prospects due to the need for more information regarding the controversial roles of MSCs in crosstalk tumors. In this review, we discuss the therapeutic potential of MSCs, the controversial roles played by MSCs in cancer progression, and the anticancer therapeutic strategies that are in association with MSCs. Finally, the clinical trials designed for the direct use of MSCs for cancer therapy or for their use in decreasing the side effects of other cancer therapies are also mentioned in this review to evaluate the current status of MSC-based cancer therapies.
Collapse
Affiliation(s)
- Züleyha Baran
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Anadolu University, Eskişehir, Turkey
| | - Melisa Çetinkaya
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Yusuf Baran
- Laboratory of Cancer Genetics, Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey.
| |
Collapse
|
5
|
Manikantan V, Ripley NE, Nielsen MK, Dangoudoubiyam S. Protein profile of extracellular vesicles derived from adult Parascaris spp. Parasit Vectors 2024; 17:426. [PMID: 39390471 PMCID: PMC11468347 DOI: 10.1186/s13071-024-06502-3] [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: 07/01/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Parascaris spp. represent a significant threat to equine health worldwide, particularly in foals. The long-term survival of parasites in the host necessitates persistent modulation of the host immune response. Intercellular communication achieved through the exchange of molecules via extracellular vesicles (EVs) released from the parasite could be a crucial factor in this regard. This study aimed to isolate and characterize EVs released by adult male and female Parascaris worms and conduct a proteomic analysis to identify sex-specific proteins and potential immunomodulatory factors. METHODS Live adult Parascaris worms were collected, and EVs were isolated from spent culture media using differential ultracentrifugation. Nanoparticle tracking analysis and transmission electron microscopy confirmed the size, concentration, and morphology of the isolated EVs. Proteins within the isolated EVs were analyzed using mass spectrometry-based proteomics (LC-MS/MS). RESULTS Proteomic analysis revealed a total of 113 proteins in Parascaris EVs, with several proteins showing homology to known helminth exosome proteins and exhibiting immunomodulatory functions. Sex-specific differences in EV protein composition were observed, with a distinct abundance of C-type lectins in female EVs, suggesting potential sex-specific roles or regulation. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed metabolic pathways shared between male and female Parascaris EVs, as well as differences in signal transduction, and cell growth and death pathways, indicating sex-specific variations. CONCLUSIONS These findings imply that Parascaris EVs and their protein cargo are complex. This data potentially opens avenues for discovering innovative approaches to managing and understanding helminth infection.
Collapse
Affiliation(s)
- Vishnu Manikantan
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, 47907, USA
| | - Nichol E Ripley
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Martin K Nielsen
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40503, USA
| | - Sriveny Dangoudoubiyam
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, 47907, USA.
| |
Collapse
|
6
|
Kaplelach AK, Murchison CF, Kojima K, Mobley JA, Arrant AE. Increased levels of extracellular matrix proteins associated with extracellular vesicles from brains of aged mice. Aging Cell 2024:e14359. [PMID: 39377264 DOI: 10.1111/acel.14359] [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: 06/05/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 10/09/2024] Open
Abstract
Extracellular vesicles (EVs) are secreted by all major cell types of the brain, providing a mode of intercellular communication and a pathway for disposal of cellular debris. EVs help maintain healthy brain function, but may also contribute to diseases affecting the brain. EVs might contribute to aging of the brain, as aging-related processes such as inflammation and cellular senescence may alter EV cargo, promoting further inflammation and senescence. However, the effects of aging on brain EVs and the function of EVs in the aging brain remain poorly understood. To address this question, we measured the levels and protein cargo of EVs isolated from the brains of 4-, 12-, and 22-month-old C57BL/6J mice. We detected no changes in EV levels, but observed age-dependent changes in EV proteins. EV fractions from aged (22 month old) brains contained higher levels of extracellular matrix proteins than EV fractions from young (4 month old) brains, with intermediate levels in 12-month-old brains. Specifically, EV fractions from aged mice contained elevated levels of hyaluronan and proteoglycan link proteins 1 and 2 and several chondroitin sulfate proteoglycans (CSPGs). Analysis of extracellular matrix in several brain regions of aged mice revealed increased immunolabeling for the CSPG aggrecan, but reduced labeling with Wisteria floribunda agglutinin, which binds to chondroitin sulfate side chains of CSPGs. These data are consistent with prior studies showing changes to the composition of extracellular matrix in aged brains, and indicate a novel association of EVs with changes in the extracellular matrix of the aging brain.
Collapse
Affiliation(s)
- Azariah K Kaplelach
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charles F Murchison
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kyoko Kojima
- Institutional Research Core Program/Mass Spectrometry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James A Mobley
- Institutional Research Core Program/Mass Spectrometry, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
7
|
Huang Y, Feng J, Xu J, Dong L, Su W, Li B, Witwer KW, Zheng L. Associations of age and sex with characteristics of extracellular vesicles and protein-enriched fractions of blood plasma. Aging Cell 2024:e14356. [PMID: 39373063 DOI: 10.1111/acel.14356] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
Extracellular vesicles (EVs) are nanosized particles that are released by various cell types and play vital roles in intercellular communication. They carry biological molecules reflecting the physiological and pathological states of their source cells and tissues, showing potential as biomarkers. However, the impact of demographic factors like age and sex on the properties of blood plasma EVs remains underexplored. This study aims to fill this gap by evaluating how these factors influence the particle count and proteomic profiles of plasma EV preparations and corresponding protein fractions. Plasma samples from 120 healthy volunteers were collected and pooled into six groups: young males (age: 27.6 ± 4.0), young females (27.4 ± 3.8), middle-aged males (48.8 ± 3.8), middle-aged females (48.9 ± 3.9), old males (69.3 ± 3.9), and old females (69.4 ± 4.3). EV- and protein-enriched fractions were separated by size-exclusion chromatography (SEC). Fractions were characterized for particle number concentration and protein composition to identify characteristics affected by age and biological sex. Plasma EVs and corresponding protein fractions exhibited distinct characteristics, with differential enrichment of markers related to EVs and other blood components, including lipoproteins. Proteomic profiles of both EVs and protein fractions displayed sex- and age-dependent differences. Differentially abundant proteins displayed functions previously identified in the context of aging and sex differences, highlighting their utility as biomarkers. Age and sex significantly affect the characteristics of plasma EVs and proteins, potentially influencing their efficacy and interpretation as biomarkers in clinical applications. This study lays the groundwork for detailed mechanistic research to understand how EVs mediate age- and sex-related effects in health.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Junjie Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiannan Xu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liang Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanting Su
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Nouvel J, Quevedo GB, Prinz T, Masood R, Daaboul G, Gainey‐Schleicher T, Wittel U, Chikhladze S, Melykuti B, Helmstaedter M, Winkler K, Nazarenko I, Pütz G. Separation of small extracellular vesicles (sEV) from human blood by Superose 6 size exclusion chromatography. J Extracell Vesicles 2024; 13:e70008. [PMID: 39441012 PMCID: PMC11497763 DOI: 10.1002/jev2.70008] [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: 07/17/2023] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Extracellular vesicles (EVs) are valuable targets for liquid biopsy. However, attempts to introduce EV-based biomarkers into clinical practice have not been successful to the extent expected. One of the reasons for this failure is the lack of reliable methods for EV baseline purification from complex biofluids, such as cell-free plasma or serum. Because available one-step approaches for EV isolation are insufficient to purify EVs, the majority of studies on clinical samples were performed either on a mixture of EVs and lipoproteins, whilst the real number of EVs and their individual specific biomarker content remained elusive, or on a low number of samples of sufficient volume to allow elaborate 2-step EV separation by size and density, resulting in a high purity but utmost low recovery. Here we introduce Fast Protein Liquid Chromatography (FPLC) using Superose 6 as a matrix to obtain small EVs from biofluids that are almost free of soluble proteins and lipoproteins. Along with the estimation of a realistic number of small EVs in human samples, we show temporal resolution of the effect of the duration of postprandial phase on the proportion of lipoproteins in purified EVs, suggesting acceptable time frames additionally to the recommendation to use fasting samples for human studies. Furthermore, we assessed a potential value of pure EVs for liquid biopsy, exemplarily examining EV- and tumour-biomarkers in pure FPLC-derived fractions isolated from the serum of patients with pancreatic cancer. Consistent among different techniques, showed the presence of diseases-associated biomarkers in pure EVs, supporting the feasibility of using single-vesicle analysis for liquid biopsy.
Collapse
Affiliation(s)
- Jerome Nouvel
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| | - Gonzalo Bustos Quevedo
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Tony Prinz
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Ramsha Masood
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | | | - Tanja Gainey‐Schleicher
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Uwe Wittel
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Department of General and Visceral SurgeryFreiburgGermany
| | - Sophia Chikhladze
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Department of General and Visceral SurgeryFreiburgGermany
| | - Bence Melykuti
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
| | - Martin Helmstaedter
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- IMITATE EM Core FacilityFreiburgGermany
| | - Karl Winkler
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital EpidemiologyFreiburgGermany
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Hahn‐SchikardFreiburgGermany
- German Cancer Consortium (DKTK)Partner Site Freiburg and German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Gerhard Pütz
- Medical Center, Faculty of Medicine, University of FreiburgUniversity of FreiburgFreiburgGermany
- Institute of Clinical Chemistry and Laboratory MedicineFreiburgGermany
| |
Collapse
|
9
|
Palviainen M, Puutio J, Østergaard RH, Eble JA, Maaninka K, Butt U, Ndika J, Kari OK, Kamali‐Moghaddam M, Kjaer‐Sorensen K, Oxvig C, Aransay AM, Falcon‐Perez JM, Federico A, Greco D, Laitinen S, Hayashi Y, Siljander PR. Beyond basic characterization and omics: Immunomodulatory roles of platelet-derived extracellular vesicles unveiled by functional testing. J Extracell Vesicles 2024; 13:e12513. [PMID: 39330919 PMCID: PMC11428872 DOI: 10.1002/jev2.12513] [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: 12/05/2023] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Renowned for their role in haemostasis and thrombosis, platelets are also increasingly recognized for their contribution in innate immunity, immunothrombosis and inflammatory diseases. Platelets express a wide range of receptors, which allows them to reach a variety of activation endpoints and grants them immunomodulatory functions. Activated platelets release extracellular vesicles (PEVs), whose formation and molecular cargo has been shown to depend on receptor-mediated activation and environmental cues. This study compared the immunomodulatory profiles of PEVs generated via activation of platelets by different receptors, glycoprotein VI, C-type lectin-like receptor 2 and combining all thrombin-collagen receptors. Functional assays in vivo in zebrafish and in vitro in human macrophages highlighted distinct homing and secretory responses triggered by the PEVs. In contrast, omics analyses of protein and miRNA cargo combined with physicochemical particle characterization found only subtle differences between the activated PEV types, which were insufficient to predict their different immunomodulatory functions. In contrast, constitutively released PEVs, formed in the absence of an exogenous activator, displayed a distinct immunomodulatory profile from the receptor-induced PEVs. Our findings underscore that PEVs are tunable through receptor-mediated activation. To truly comprehend their role(s) in mediating platelet functions among immune cells, conducting functional assays is imperative.
Collapse
Affiliation(s)
- Mari Palviainen
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Johanna Puutio
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | | | - Johannes A. Eble
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MünsterMünsterGermany
| | - Katariina Maaninka
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Umar Butt
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Joseph Ndika
- Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Otto K. Kari
- Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Masood Kamali‐Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life LaboratoryUppsala UniversityUppsalaSweden
| | | | - Claus Oxvig
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Ana M. Aransay
- Genome Analysis Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology Alliance (BRTA)MendaroSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd)MadridSpain
| | - Juan M. Falcon‐Perez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd)MadridSpain
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA)DerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE); Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE); Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Division of Pharmaceutical Biosciences, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
| | - Saara Laitinen
- Research and DevelopmentFinnish Red Cross Blood Service (FRCBS)HelsinkiFinland
| | - Yuya Hayashi
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusDenmark
| | - Pia R.‐M. Siljander
- EV Group, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, and CURED, Drug Research Program, Faculty of PharmacyUniversity of HelsinkiHelsinkiFinland
- EV Core, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
10
|
Vahab SA, V VK, Kumar VS. Exosome-based drug delivery systems for enhanced neurological therapeutics. Drug Deliv Transl Res 2024:10.1007/s13346-024-01710-x. [PMID: 39325272 DOI: 10.1007/s13346-024-01710-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are small extracellular vesicles naturally secreted by cells into body fluids, enriched with bioactive molecules such as RNAs, proteins, and lipids. These nanosized vesicles play a crucial role in physiological and pathological processes by facilitating intercellular communication and modulating cellular responses, particularly within the central nervous system (CNS). Their ability to cross the blood-brain barrier and reflect the characteristics of their parent cells makes exosomal cargo a promising candidate for biomarkers in the early diagnosis and clinical assessment of neurological conditions. This review offers a comprehensive overview of current knowledge on the characterization of mammalian-derived exosomes, their application as drug delivery systems for neurological disorders, and ongoing clinical trials involving exosome-loaded cargo. Despite their promising attributes, a significant challenge remains the lack of standardized isolation methods, as current techniques are often complex, costly, and require sophisticated equipment, affecting the scalability and affordability of exosome-based therapies. The review highlights the engineering potential of exosomes, emphasizing their ability to be customized for targeted therapeutic delivery through surface modification or conjugation. Future advancements in addressing these challenges and leveraging the unique properties of exosomes could lead to innovative and effective therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Safa A Vahab
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India
| | - Vyshma K V
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India
| | - Vrinda S Kumar
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi-682041, Kerala, India.
| |
Collapse
|
11
|
Mao Y, Suryawanshi A, Patial S, Saini Y. Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Front Pharmacol 2024; 15:1460692. [PMID: 39386033 PMCID: PMC11461968 DOI: 10.3389/fphar.2024.1460692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Exosomes, membrane-bound extracellular vesicles, ranging from approximately 30-200 nm in diameter, are released by almost all cell types and play critical roles in intercellular communication. In response to the prevailing stress, the exosome-bound protein signatures vary in abundance and composition. To identify the bronchoalveolar lavage fluid (BALF) exosome-bound proteins associated with mucoinflammatory lung disease and to gain insights into their functional implications, we compared BALF exosomes-derived proteins from adult Scnn1b transgenic (Scnn1b-Tg+) and wild type (WT) mice. A total of 3,144 and 3,119 proteins were identified in BALF exosomes from Scnn1b-Tg+ and WT mice, respectively. Using cutoff criteria (Log2 fold-change > 1 and adjusted p-value < 0.05), the comparison of identified proteins revealed 127 and 30 proteins that were significantly upregulated and downregulated, respectively, in Scnn1b-Tg+ versus WT mice. In addition, 52 and 27 proteins were exclusively enriched in Scnn1b-Tg+ and WT mice, respectively. The identified exosome-bound proteins from the homeostatic airspaces of WT mice were mostly relevant to the normal physiological processes. The protein signatures enriched in the BALF exosomes of Scnn1b-Tg+ mice were relevant to macrophage activation and mucoinflammatory processes. Ingenuity pathway analyses revealed that the enriched proteins in Scnn1b-Tg+ mice contributed to the inflammatory responses and antimicrobial defense pathways. Selective proteins including, RETNLA/FIZZ1, LGALS3/Galectin-3, S100A8/MRP8, and CHIL3/YM1 were immunolocalized to specific cell types. The comparative analysis between enriched BALF exosome proteins and previously identified differentially upregulated genes in Scnn1b-Tg+ versus WT mice suggested that the compartment-/cell-specific upregulation in gene expression dictates the enrichment of their respective proteins in the lung airspaces. Taken together, this study demonstrates that the BALF exosome-bound protein signatures reflect disease-relevant disturbances. Our findings suggest that the exosomes carry disease-relevant protein signatures that can be used as a diagnostic as well as predictive biomarkers for mucoinflammatory lung disease.
Collapse
Affiliation(s)
- Yun Mao
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sonika Patial
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
12
|
Bahram Sangani N, Koetsier J, Gomes AR, Diogo MM, Fernandes TG, Bouwman FG, Mariman ECM, Ghazvini M, Gribnau J, Curfs LMG, Reutelingsperger CP, Eijssen LMT. Involvement of extracellular vesicle microRNA clusters in developing healthy and Rett syndrome brain organoids. Cell Mol Life Sci 2024; 81:410. [PMID: 39305343 PMCID: PMC11416455 DOI: 10.1007/s00018-024-05409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 09/25/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by de novo mutations in the MECP2 gene. Although miRNAs in extracellular vesicles (EVs) have been suggested to play an essential role in several neurological conditions, no prior study has utilized brain organoids to profile EV-derived miRNAs during normal and RTT-affected neuronal development. Here we report the spatiotemporal expression pattern of EV-derived miRNAs in region-specific forebrain organoids generated from female hiPSCs with a MeCP2:R255X mutation and the corresponding isogenic control. EV miRNA and protein expression profiles were characterized at day 0, day 13, day 40, and day 75. Several members of the hsa-miR-302/367 cluster were identified as having a time-dependent expression profile with RTT-specific alterations at the latest developmental stage. Moreover, the miRNA species of the chromosome 14 miRNA cluster (C14MC) exhibited strong upregulation in RTT forebrain organoids irrespective of their spatiotemporal location. Together, our results suggest essential roles of the C14MC and hsa-miR-302/367 clusters in EVs during normal and RTT-associated neurodevelopment, displaying promising prospects as biomarkers for monitoring RTT progression.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Ana Rita Gomes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Freek G Bouwman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mehrnaz Ghazvini
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Joost Gribnau
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Developmental Biology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands.
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands.
| | - Lars M T Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- Department of Bioinformatics-BiGCaT, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
13
|
Lu P, Yang L, Chen W, Li K, Chen X, Qu S. Four-dimensional trapped ion mobility spectrometry proteomics reveals circulating extracellular vesicles encapsulated drivers of nasopharyngeal carcinoma distant dissemination. Talanta 2024; 282:126907. [PMID: 39341061 DOI: 10.1016/j.talanta.2024.126907] [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: 08/17/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck cancer with a high propensity for early metastatic spread. Emerging evidence shows that extracellular vesicles (EVs) are key players in cancer metastasis, but their role in NPC metastasis remains poorly understood. We here present the first description of the proteomic and functional profiles of serum-derived circulating small EVs in metastatic NPC patients. To enhance the capture of low-abundance signaling proteins in EVs, timsTOF-based four-dimensional label-free quantitative proteomics was employed. We found that metastatic NPC patients (M-NPC-EVs) exhibited the highest serum EV levels compared to locoregional patients (L-NPC-EVs) and healthy subjects (Normal-EVs). The proteome of M-NPC-EVs differed substantially from L-NPC-EVs and was functionally enriched in pathways regulating cell polarity and motility, glucose metabolism, and angiogenesis. Functional assays testing individual EV samples demonstrated that M-NPC-EVs pronouncedly enhanced NPC cell migration, invasion, and the formation of lamellipodia and filopodia in vitro, and promoted angiogenesis in subcutaneous Matrigel plugs in vivo. In silico analyses suggested that PTPRA, TPI1 and GPI highly enriched in M-NPC-EVs were putative drivers underlying the motogenic and angiogenic activities of M-NPC-EVs, and their high expression levels were associated with a poor prognosis of NPC patients. The increased expression of PTPRA, TPI1 and GPI in M-NPC-EVs was then validated in an independent cohort consisting of 175 NPC patients (locoregional n = 114; metastatic n = 61). Together, utilizing patient-derived EVs, we mimicked the potential pro-metastatic functions of EVs in NPC patients in vitro and in vivo and provided novel insights into their bioactive cargoes.
Collapse
Affiliation(s)
- Pingan Lu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Liu Yang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Weiling Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Kaiguo Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Xuxia Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, 530021, Guangxi Autonomous Region, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China; Guangxi Key Laboratory of High-Incidence Tumor Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China; Guangxi Nasopharyngeal Carcinoma Clinical Research Center, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
14
|
Stephens KK, Finnerty RM, Grant DG, Winuthayanon S, Martin-DeLeon PA, Winuthayanon W. Proteomic analysis and in vivo visualization of extracellular vesicles from mouse oviducts during pre-implantation embryo development. FASEB J 2024; 38:e70035. [PMID: 39239798 PMCID: PMC11384279 DOI: 10.1096/fj.202400041rr] [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: 01/06/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Pre-implantation embryonic development occurs in the oviduct during the first few days of pregnancy. The presence of oviductal extracellular vesicles (oEVs, also called oviductosomes) is crucial for pre-implantation embryonic development in vivo as oEVs often contain molecular transmitters such as proteins. Therefore, evaluating oEV cargo during early pregnancy could provide insights into factors required for proper early embryonic development that are missing in the current in vitro embryo culture setting. In this study, we isolated oEVs from the oviductal fluid at estrus and different stages of early embryonic development. The 2306-3066 proteins in oEVs identified at the different time points revealed 58-60 common EV markers identified in exosome databases. Oviductal extracellular vesicle proteins from pregnant samples significantly differed from those in non-pregnant samples. In addition, superovulation changes the protein contents in oEVs compared to natural ovulation at estrus. Importantly, we have identified that embryo-protectant proteins such as high-mobility protein group B1 and serine (or cysteine) peptidase inhibitor were only enriched in the presence of embryos. We also visualized the physical interaction of EVs and the zona pellucida of 4- to 8-cell stage embryos using transmission electron microscopy as well as in vivo live imaging of epithelial cell-derived GFP-tagged CD9 mouse model. All protein data in this study are readily available to the scientific community in a searchable format at https://genes.winuthayanon.com/winuthayanon/oviduct_ev_proteins/. In conclusion, we identified oEVs proteins that could be tested to determine whether they can improve embryonic developmental outcomes in vivo and in vitro setting.
Collapse
Affiliation(s)
- Kalli K. Stephens
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
| | - Ryan M. Finnerty
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
- Translational Biosciences Program, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - DeAna G. Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, MO, 65211, USA
| | - Sarayut Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
| | | | - Wipawee Winuthayanon
- Division of Animal Sciences, College of Agriculture, Food and Natural Resources, University of Missouri, MO, 65201, USA
- Department of OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
- Translational Biosciences Program, School of Medicine, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
15
|
Di Bella MA, Taverna S. Extracellular Vesicles: Diagnostic and Therapeutic Applications in Cancer. BIOLOGY 2024; 13:716. [PMID: 39336143 PMCID: PMC11446462 DOI: 10.3390/biology13090716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
In recent years, knowledge of cell-released extracellular vesicle (EV) functions has undergone rapid growth. EVs are membrane vesicles loaded with proteins, nucleic acids, lipids, and bioactive molecules. Once released into the extracellular space, EVs are delivered to target cells that may go through modifications in physiological or pathological conditions. EVs are nano shuttles with a crucial role in promoting short- and long-distance cell-cell communication. Comprehension of the mechanism that regulates this process is a benefit for both medicine and basic science. Currently, EVs attract immense interest in precision and nanomedicine for their potential use in diagnosis, prognosis, and therapies. This review reports the latest advances in EV studies, focusing on the nature and features of EVs and on conventional and emerging methodologies used for their separation, characterization, and visualization. By searching an extended portion of the relevant literature, this work aims to give a summary of advances in nanomedical applications of EVs. Moreover, concerns that require further studies before translation to clinical applications are discussed.
Collapse
Affiliation(s)
- Maria Antonietta Di Bella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 90146 Palermo, Italy
| |
Collapse
|
16
|
Lin S, Chang Y, Lee W, Chiang C, Liu S, Lee H, Jeng L, Shyu W. Role of STAT3-FOXO3 Signaling in the Modulation of Neuroplasticity by PD-L1-HGF-Decorated Mesenchymal Stem Cell-Derived Exosomes in a Murine Stroke Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404882. [PMID: 39049677 PMCID: PMC11423231 DOI: 10.1002/advs.202404882] [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: 05/08/2024] [Revised: 06/28/2024] [Indexed: 07/27/2024]
Abstract
The limited therapeutic strategies available for stroke leave many patients disabled for life. This study assessed the potential of programmed death-ligand 1 (PD-L1) and hepatocyte growth factor (HGF)-engineered mesenchymal stem cell-derived exosomes (EXO-PD-L1-HGF) in enhancing neurological recovery post-stroke. EXO-PD-L1-HGF, which efficiently endocytosed into target cells, significantly diminishes the H2O2-induced neurotoxicity and increased the antiapoptotic proteins in vitro. EXO-PD-L1-HGF attenuates inflammation by inhibiting T-cell proliferation and increasing the number of CD8+CD122+IL-10+ regulatory T cells. Intravenous injection of EXO-PD-L1-HGF could target stromal cell-derived factor-1α (SDF-1α+) cells over the peri-infarcted area of the ischemic brain through CXCR4 upregulation and accumulation in neuroglial cells post-stroke. EXO-PD-L1-HGF facilitates endogenous nestin+ neural progenitor cell (NPC)-induced neurogenesis via STAT3-FOXO3 signaling cascade, which plays a pivotal role in cell survival and neuroprotection, thereby mitigating infarct size and enhancing neurological recovery in a murine stroke model. Moreover, increasing populations of the immune-regulatory CD19+IL-10+ and CD8+CD122+IL-10+ cells, together with reducing populations of proinflammatory cells, created an anti-inflammatory microenvironment in the ischemic brain. Thus, innovative approaches employing EXO-PD-L1-HGF intervention, which targets SDF-1α+ expression, modulates the immune system, and enhances the activation of resident nestin+ NPCs, might significantly alter the brain microenvironment and create a niche conducive to inducing neuroplastic regeneration post-stroke.
Collapse
Affiliation(s)
- Syuan‐Ling Lin
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
| | - Yi‐Wen Chang
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Department of Medical ResearchNational Taiwan University HospitalTaipei100Taiwan
| | - Wei Lee
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
| | - Chih‐Sheng Chiang
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
| | - Shih‐Ping Liu
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
| | - Hsu‐Tung Lee
- Graduate Institute of Medical SciencesNational Defense Medical CenterTaipei114Taiwan
- Department of Post‐Baccalaureate Medicine, College of MedicineNational Chung Hsing UniversityTaichung402Taiwan
- Division of neurosurgical Oncology Neurological InstituteTaichung Veterans General HospitalTaichung407Taiwan
| | - Long‐Bin Jeng
- Cell Therapy CenterChina Medical University HospitalTaichung404Taiwan
- Organ Transplantation CenterChina Medical University HospitalTaichung404Taiwan
| | - Woei‐Cherng Shyu
- Translational Medicine Research Center and Department of NeurologyChina Medical University HospitalTaichung404Taiwan
- Graduate Institute of Biomedical Sciences and New Drug Development CenterChina Medical UniversityTaichung404Taiwan
- Department of Occupational TherapyAsia UniversityTaichung413Taiwan
| |
Collapse
|
17
|
Parvin A, Erabi G, Mohammadpour D, Maleki-Kakelar H, Sadeghpour S, Pashaei MR, Taheri-Anganeh M, Ghasemnejad-Berenji H. Infertility: Focus on the therapeutic potential of extracellular vesicles. Reprod Biol 2024; 24:100925. [PMID: 39018753 DOI: 10.1016/j.repbio.2024.100925] [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: 03/30/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Infertility is a well-known problem that arises from a variety of reproductive diseases. Until now, researchers have tried various methods to restore fertility, including medication specific to the cause, hormone treatments, surgical removals, and assisted reproductive technologies. While these methods do produce results, they do not consistently lead to fertility restoration in every instance. The use of exosome therapy has significant potential in treating infertility in patients. This is because exosomes, microvesicles, and apoptotic bodies, which are different types of vesicles, play a crucial role in transferring bioactive molecules that aid in cell-to-cell communication. Reproductive fluids can transport a variety of molecular cargos, such as miRNAs, mRNAs, proteins, lipids, and DNA molecules. The percentage of these cargos in the fluids can be linked to their physiological and pathological status. EVs are involved in several physiological and pathological processes and offer interesting non-cellular therapeutic possibilities to treat infertility. EVs (extracellular vesicles) transplantation has been shown in many studies to be a key part of regenerating different parts of the reproductive system, including the production of oocytes and the start of sperm production. Nevertheless, the existing evidence necessitates testifying to the effectiveness of injecting EVs in resolving reproductive problems among humans. This review focuses on the current literature about infertility issues in both females and males, specifically examining the potential treatments involving extracellular vesicles (EVs).
Collapse
Affiliation(s)
- Ali Parvin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Donna Mohammadpour
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Obstetrics & Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaei
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
18
|
Zhou H, Wu W, Zhang Q, Zhang T, Jiang S, Chang L, Xie Y, Zhu J, Zhou D, Zhang Y, Xu P. Proteome overview of exosome derived from plasma of cows infected with Mycobacterium bovis. Tuberculosis (Edinb) 2024; 148:102541. [PMID: 39002312 DOI: 10.1016/j.tube.2024.102541] [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: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
Bovine tuberculosis (bTB), primarily caused by Mycobacterium bovis (M. bovis), is a globally zoonotic disease with significant economic impacts. Plasma exosomes have been extensively used for investigating disease processes and exploring biomarkers. While mass spectrometry (MS)-based proteomic analysis of plasma exosomes has been employed for human tuberculosis (TB) studies, it has not yet been applied to bTB. Therefore, a comprehensive proteomic overview of plasma exosomes from M. bovis-infected cows is essential. In this study, we presented an extensive proteomic analysis of plasma exosomes from 89 M. bovis-infected cows across three farms, using data dependent acquisition (DDA) mode. Our analysis encompasses 239,894 spectra, 6,011 peptides and 835 proteins. The proteomic overview revealed both consistencies and differences among individual cows, supplements 595 proteins to the bovine exosome library, and enriches tuberculosis and related pathways. Additionally, six pathways were validated as immune response pathways, and three proteins (CATHL1, H1-1, and LCN2) were identified as potential indicators of bTB. This study is the first to investigate the exosome proteome of plasma from cows infected with M. bovis, providing a valuable dataset for exploring candidate bTB markers and understanding the mechanisms of host defense against M. bovis.
Collapse
Affiliation(s)
- Hangfan Zhou
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Wenhui Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Second Clinical Medicine Collage, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qilong Zhang
- Beijing Center for Animal Disease Control and Prevention, Beijing, 102629, China
| | - Tao Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Songhao Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Lei Chang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Yuping Xie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jiaqiang Zhu
- Beijing Xinhui Purui Technology Development Co., Ltd, Beijing, 102200, China
| | - Degang Zhou
- Beijing Center for Animal Disease Control and Prevention, Beijing, 102629, China.
| | - Yao Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Ping Xu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Beijing Institute of Lifeomics, Beijing, 102206, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Second Clinical Medicine Collage, Guangzhou Higher Education Mega Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, 110122, China; School of Basic Medicine, Anhui Medical University, Hefei, 230032, China; Department of Biomedicine, Medical College, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
19
|
Zhang Y, Yu L, Yang M, Han B, Luo J, Jing R. Model fusion for predicting unconventional proteins secreted by exosomes using deep learning. Proteomics 2024; 24:e2300184. [PMID: 38643383 DOI: 10.1002/pmic.202300184] [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: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/22/2024]
Abstract
Unconventional secretory proteins (USPs) are vital for cell-to-cell communication and are necessary for proper physiological processes. Unlike classical proteins that follow the conventional secretory pathway via the Golgi apparatus, these proteins are released using unconventional pathways. The primary modes of secretion for USPs are exosomes and ectosomes, which originate from the endoplasmic reticulum. Accurate and rapid identification of exosome-mediated secretory proteins is crucial for gaining valuable insights into the regulation of non-classical protein secretion and intercellular communication, as well as for the advancement of novel therapeutic approaches. Although computational methods based on amino acid sequence prediction exist for predicting unconventional proteins secreted by exosomes (UPSEs), they suffer from significant limitations in terms of algorithmic accuracy. In this study, we propose a novel approach to predict UPSEs by combining multiple deep learning models that incorporate both protein sequences and evolutionary information. Our approach utilizes a convolutional neural network (CNN) to extract protein sequence information, while various densely connected neural networks (DNNs) are employed to capture evolutionary conservation patterns.By combining six distinct deep learning models, we have created a superior framework that surpasses previous approaches, achieving an ACC score of 77.46% and an MCC score of 0.5406 on an independent test dataset.
Collapse
Affiliation(s)
- Yonglin Zhang
- Department of Clinical Pharmacy and Pharmacy Management, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lezheng Yu
- School of Chemistry and Materials Science, Guizhou Education University, Guiyang, Guizhou, China
| | - Ming Yang
- Department of Clinical Pharmacy and Pharmacy Management, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Bin Han
- GCP Center/Institute of Drug Clinical Trials, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jiesi Luo
- Basic Medical College, Southwest Medical University, Luzhou, Sichuan, China
| | - Runyu Jing
- School of Cyber Science and Engineering, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Useckaite Z. Extracellular vesicles: A potential new way to assess cholestasis. Obstet Med 2024; 17:179-183. [PMID: 39262910 PMCID: PMC11384818 DOI: 10.1177/1753495x241264325] [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] [Accepted: 06/04/2024] [Indexed: 09/13/2024] Open
Abstract
Extracellular vesicles (EVs) are small, nonreplicating, lipid-encapsulated nanoparticles that carry protein and nucleic acid cargo derived from their tissue of origin. Due to their capacity to provide comparable insights to solid organ biopsy through a minimally invasive collection procedure, EVs provide an attractive biomarker source. This review will provide an insight, how EVs in circulation may provide a novel way to assess cholestasis and will address the possibility of getting a better understanding of the mechanisms of cholestasis of pregnancy through the use of serial hepatic-specific EVs as a window.
Collapse
Affiliation(s)
- Zivile Useckaite
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Australia
| |
Collapse
|
21
|
Beutgen VM, Shinkevich V, Pörschke J, Meena C, Steitz AM, Pogge von Strandmann E, Graumann J, Gómez-Serrano M. Secretome Analysis Using Affinity Proteomics and Immunoassays: A Focus on Tumor Biology. Mol Cell Proteomics 2024; 23:100830. [PMID: 39147028 PMCID: PMC11417252 DOI: 10.1016/j.mcpro.2024.100830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/20/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
The study of the cellular secretome using proteomic techniques continues to capture the attention of the research community across a broad range of topics in biomedical research. Due to their untargeted nature, independence from the model system used, historically superior depth of analysis, as well as comparative affordability, mass spectrometry-based approaches traditionally dominate such analyses. More recently, however, affinity-based proteomic assays have massively gained in analytical depth, which together with their high sensitivity, dynamic range coverage as well as high throughput capabilities render them exquisitely suited to secretome analysis. In this review, we revisit the analytical challenges implied by secretomics and provide an overview of affinity-based proteomic platforms currently available for such analyses, using the study of the tumor secretome as an example for basic and translational research.
Collapse
Affiliation(s)
- Vanessa M Beutgen
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany; Core Facility Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
| | - Veronika Shinkevich
- Institute of Pharmacology, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
| | - Johanna Pörschke
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Celina Meena
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Anna M Steitz
- Translational Oncology Group, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany; Core Facility Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany.
| | - María Gómez-Serrano
- Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
| |
Collapse
|
22
|
Zhuang T, Wang S, Yu X, He X, Guo H, Ou C. Current status and future perspectives of platelet-derived extracellular vesicles in cancer diagnosis and treatment. Biomark Res 2024; 12:88. [PMID: 39183323 PMCID: PMC11346179 DOI: 10.1186/s40364-024-00639-0] [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: 06/29/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Platelets are a significant component of the cell population in the tumour microenvironment (TME). Platelets influence other immune cells and perform cross-talk with tumour cells, playing an important role in tumour development. Extracellular vesicles (EVs) are small membrane vesicles released from the cells into the TME. They can transfer biological information, including proteins, nucleic acids, and metabolites, from secretory cells to target receptor cells. This process affects the progression of various human diseases, particularly cancer. In recent years, several studies have demonstrated that platelet-derived extracellular vesicles (PEVs) can help regulate the malignant biological behaviours of tumours, including malignant proliferation, resistance to cell death, invasion and metastasis, metabolic reprogramming, immunity, and angiogenesis. Consequently, PEVs have been identified as key regulators of tumour progression. Therefore, targeting PEVs is a potential strategy for tumour treatment. Furthermore, the extensive use of nanomaterials in medical research has indicated that engineered PEVs are ideal delivery systems for therapeutic drugs. Recent studies have demonstrated that PEV engineering technologies play a pivotal role in the treatment of tumours by combining photothermal therapy, immunotherapy, and chemotherapy. In addition, aberrant changes in PEVs are closely associated with the clinicopathological features of patients with tumours, which may serve as liquid biopsy markers for early diagnosis, monitoring disease progression, and the prognostic assessment of patients with tumours. A comprehensive investigation into the role and potential mechanisms of PEVs in tumourigenesis may provide novel diagnostic biomarkers and potential therapeutic strategies for treating human tumours.
Collapse
Affiliation(s)
- Tongtao Zhuang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shenrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoqian Yu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hongbin Guo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
23
|
Zertuche-Martínez C, Velázquez-Enríquez JM, González-García K, Santos-Álvarez JC, Romero-Tlalolini MDLÁ, Pina-Canseco S, Pérez-Campos Mayoral L, Muriel P, Villa-Treviño S, Baltiérrez-Hoyos R, Arellanes-Robledo J, Vásquez-Garzón VR. Discovery of candidate biomarkers from plasma-derived extracellular vesicles of patients with cirrhosis and hepatocellular carcinoma: an exploratory proteomic study. Mol Omics 2024; 20:483-495. [PMID: 39011654 DOI: 10.1039/d4mo00043a] [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: 07/17/2024]
Abstract
Extracellular vesicles (EVs) represent an attractive source of biomarkers due to their biomolecular cargo. The aim of this study was to identify candidate protein biomarkers from plasma-derived EVs of patients with liver cirrhosis (LC) and hepatocellular carcinoma (HCC). Plasma-derived EVs from healthy participants (HP), LC, and HCC patients (eight samples each) were subjected to label-free quantitative proteomic analysis using LC-MS/MS. A total of 248 proteins were identified, and differentially expressed proteins (DEPs) were obtained after pairwise comparison. We found that DEPs mainly involve complement cascade activation, coagulation pathways, cholesterol metabolism, and extracellular matrix components. By choosing a panel of up- and down-regulated proteins involved in cirrhotic and carcinogenesis processes, TGFBI, LGALS3BP, C7, SERPIND1, and APOC3 were found to be relevant for LC patients, while LRG1, TUBA1C, TUBB2B, ACTG1, C9, HP, FGA, FGG, FN1, PLG, APOB and ITIH2 were associated with HCC patients, which could discriminate both diseases. In addition, we identified the top shared proteins in both diseases, which included LCAT, SERPINF2, A2M, CRP, and VWF. Thus, our exploratory proteomic study revealed that these proteins might play an important role in the disease progression and represent a panel of candidate biomarkers for the prognosis and diagnosis of LC and HCC.
Collapse
Affiliation(s)
- Cecilia Zertuche-Martínez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Karina González-García
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | | | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina UNAM UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Laura Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07000, Mexico
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07360, Mexico
| | - Rafael Baltiérrez-Hoyos
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico.
| | | | - Verónica Rocío Vásquez-Garzón
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico.
| |
Collapse
|
24
|
Wang Z, Zhou X, Kong Q, He H, Sun J, Qiu W, Zhang L, Yang M. Extracellular Vesicle Preparation and Analysis: A State-of-the-Art Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401069. [PMID: 38874129 PMCID: PMC11321646 DOI: 10.1002/advs.202401069] [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: 01/29/2024] [Revised: 04/11/2024] [Indexed: 06/15/2024]
Abstract
In recent decades, research on Extracellular Vesicles (EVs) has gained prominence in the life sciences due to their critical roles in both health and disease states, offering promising applications in disease diagnosis, drug delivery, and therapy. However, their inherent heterogeneity and complex origins pose significant challenges to their preparation, analysis, and subsequent clinical application. This review is structured to provide an overview of the biogenesis, composition, and various sources of EVs, thereby laying the groundwork for a detailed discussion of contemporary techniques for their preparation and analysis. Particular focus is given to state-of-the-art technologies that employ both microfluidic and non-microfluidic platforms for EV processing. Furthermore, this discourse extends into innovative approaches that incorporate artificial intelligence and cutting-edge electrochemical sensors, with a particular emphasis on single EV analysis. This review proposes current challenges and outlines prospective avenues for future research. The objective is to motivate researchers to innovate and expand methods for the preparation and analysis of EVs, fully unlocking their biomedical potential.
Collapse
Affiliation(s)
- Zesheng Wang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Qinglong Kong
- The Second Department of Thoracic SurgeryDalian Municipal Central HospitalDalian116033P. R. China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Wenting Qiu
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
25
|
Dainiak N. Biology of Exfoliation of Plasma Membrane-Derived Vesicles and the Radiation Response: Historical Background, Applications in Biodosimetry and Cell-Free Therapeutics, and Quantal Mechanisms for Their Release and Function with Implications for Space Travel. Radiat Res 2024; 202:328-354. [PMID: 38981604 DOI: 10.1667/rade-24-00078.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/09/2024] [Indexed: 07/11/2024]
Abstract
This historical review of extracellular vesicles in the setting of exposure to ionizing radiation (IR) traces our understanding of how vesicles were initially examined and reported in the literature in the late 1970s (for secreted exosomes) and early 1980s (for plasma membrane-derived, exfoliated vesicles) to where we are now and where we may be headed in the next decade. An emphasis is placed on biophysical properties of extracellular vesicles, energy consumption and the role of vesiculation as an essential component of membrane turnover. The impact of intercellular signal trafficking by vesicle surface and intra-vesicular lipids, proteins, nucleic acids and metabolites is reviewed in the context of biomarkers for estimating individual radiation dose after exposure to radiation, pathogenesis of disease and development of cell-free therapeutics. Since vesicles express both growth stimulatory and inhibitory molecules, a hypothesis is proposed to consider superposition in a shared space and entanglement of molecules by energy sources that are external to human cells. Implications of this approach for travel in deep space are briefly discussed in the context of clinical disorders that have been observed after space travel.
Collapse
Affiliation(s)
- Nicholas Dainiak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
26
|
Ikhlef L, Ratti N, Durand S, Formento R, Daverat H, Boutaud M, Guillou C, Dmytruk N, Gachard N, Cosette P, Jauberteau MO, Gallet PF. Extracellular vesicles from type-2 macrophages increase the survival of chronic lymphocytic leukemia cells ex vivo. Cancer Gene Ther 2024; 31:1164-1176. [PMID: 38918490 PMCID: PMC11327105 DOI: 10.1038/s41417-024-00802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
The resistance of Chronic Lymphocytic Leukemia (CLL) B-cells to cell death is mainly attributed to interactions within their microenvironment, where they interact with various types of cells. Within this microenvironment, CLL-B-cells produce and bind cytokines, growth factors, and extracellular vesicles (EVs). In the present study, EVs purified from nurse-like cells and M2-polarized THP1 cell (M2-THP1) cultures were added to CLL-B-cells cultures. EVs were rapidly internalized by B-cells, leading to a decrease in apoptosis (P = 0.0162 and 0.0469, respectively) and an increased proliferation (P = 0.0335 and 0.0109). Additionally, they induced an increase in the resistance of CLL-B-cells to Ibrutinib, the Bruton kinase inhibitor in vitro (P = 0.0344). A transcriptomic analysis showed an increase in the expression of anti-apoptotic gene BCL-2 (P = 0.0286) but not MCL-1 and an increase in the expression of proliferation-inducing gene APRIL (P = 0.0286) following treatment with EVs. Meanwhile, an analysis of apoptotic protein markers revealed increased amounts of IGFBP-2 (P = 0.0338), CD40 (P = 0.0338), p53 (P = 0.0219) and BCL-2 (P = 0.0338). Finally, exploration of EVs protein content by mass spectrometry revealed they carry various proteins involved in known oncogenic pathways and the RNAseq analysis of CLL-B-cells treated or not with NLCs EVs show various differentially expressed genes.
Collapse
Affiliation(s)
- Léa Ikhlef
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Nina Ratti
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | | | - Rémy Formento
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Héloïse Daverat
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Marie Boutaud
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
| | - Clément Guillou
- PISSARO Proteomics Platform, Mont-Saint-Aignan Campus, Mont-Saint-Aignan, France
| | - Natalya Dmytruk
- Department of Clinical Hematology, University Hospital of Limoges, Limoges, France
| | - Nathalie Gachard
- Hematology laboratory, UMR CNRS7276/ INSERM 1262, University Hospital of Limoges, Limoges, France
| | - Pascal Cosette
- Polymers, Biopolymers, Surface Laboratory, UMR 6270 CNRS, Normandie University, UNIROUEN, INSA Rouen, Mont-Saint-Aignan, France
- HeRacLeS-PISSARO, INSERM US 51, CNRS UAR 2026, Normandie University, Mont-Saint-Aignan, France
| | - Marie-Odile Jauberteau
- University of Limoges, UMR INSERM 1308, CAPTuR, Limoges, France
- Immunology laboratory, University Hospital of Limoges, Limoges, France
| | | |
Collapse
|
27
|
Huang S, Lin J, Han X. Extracellular vesicles-Potential link between periodontal disease and diabetic complications. Mol Oral Microbiol 2024; 39:225-239. [PMID: 38227219 DOI: 10.1111/omi.12449] [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: 06/28/2023] [Revised: 12/06/2023] [Accepted: 12/25/2023] [Indexed: 01/17/2024]
Abstract
It has long been suggested that a bidirectional impact exists between periodontitis and diabetes. Periodontitis may affect diabetes glycemic control, insulin resistance, and diabetic complications. Diabetes can worsen periodontitis by delaying wound healing and increasing the chance of infection. Extracellular vesicles (EVs) are heterogeneous particles of membrane-enclosed spherical structure secreted by eukaryotes and prokaryotes and play a key role in a variety of diseases. This review will introduce the biogenesis, release, and biological function of EVs from a microbial and host cell perspective, discuss the functional properties of EVs in the development of periodontitis and diabetes, and explore their role in the pathogenesis and clinical application of these two diseases. Their clinical implication and diagnostic value are also discussed.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|
28
|
Tamimi A, Javid M, Sedighi-Pirsaraei N, Mirdamadi A. Exosome prospects in the diagnosis and treatment of non-alcoholic fatty liver disease. Front Med (Lausanne) 2024; 11:1420281. [PMID: 39144666 PMCID: PMC11322140 DOI: 10.3389/fmed.2024.1420281] [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: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
The growing prevalence of NAFLD and its global health burden have provoked considerable research on possible diagnostic and therapeutic options for NAFLD. Although various pathophysiological mechanisms and genetic factors have been identified to be associated with NAFLD, its treatment remains challenging. In recent years, exosomes have attracted widespread attention for their role in metabolic dysfunctions and their efficacy as pathological biomarkers. Exosomes have also shown tremendous potential in treating a variety of disorders. With increasing evidence supporting the significant role of exosomes in NAFLD pathogenesis, their theragnostic potential has become a point of interest in NAFLD. Expectedly, exosome-based treatment strategies have shown promise in the prevention and amelioration of NAFLD in preclinical studies. However, there are still serious challenges in preparing, standardizing, and applying exosome-based therapies as a routine clinical option that should be overcome. Due to the great potential of this novel theragnostic agent in NAFLD, further investigations on their safety, clinical efficacy, and application standardization are highly recommended.
Collapse
|
29
|
Badrhan S, Karanwal S, Pal A, Chera JS, Chauhan V, Patel A, Bhakat M, Datta TK, Kumar R. Differential protein repertoires related to sperm function identified in extracellular vesicles (EVs) in seminal plasma of distinct fertility buffalo ( Bubalus bubalis) bulls. Front Cell Dev Biol 2024; 12:1400323. [PMID: 39135778 PMCID: PMC11318068 DOI: 10.3389/fcell.2024.1400323] [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: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Buffalo bulls are backbone of Indian dairy industry, and the quality of semen donating bulls determine the overall production efficiency of dairy farms. Seminal plasma harbor millions of lipid bilayer nanovesicles known as extracellular vesicles (EVs). These EVs carry a heterogenous cargo of essential biomolecules including fertility-associated proteins which contribute to fertilizing potential of spermatozoa. In this study, we explored size, concentration, and complete proteome profiles of SP EVs from two distinct fertility groups to uncover proteins influencing bull fertility. Through Dynamic Light Scattering (DLS) it was found that purified EVs were present in 7-14 size exclusion chromatographic (SEC) fractions with sizes ranging from 146.5 to 258.7 nm in high fertile (HF) and low fertile (LF) bulls. Nanoparticle Tracking Analysis (NTA) confirmed the size of seminal EVs up to 200 nm, and concentrations varying from 2.84 to 6.82 × 1011 and 3.57 to 7.74 × 1011 particles per ml in HF and LF bulls, respectively. No significant difference was observed in size and concentration of seminal EVs between two groups. We identified a total of 1,862 and 1,807 proteins in seminal EVs of HF and LF bulls, respectively using high throughput LC-MS/MS approach. Out of these total proteins, 1,754 proteins were common in both groups and about 87 proteins were highly abundant in HF group while 1,292 were less abundant as compared to LF bulls. Gene ontology (GO) analysis, revealed that highly abundant proteins in HF group were mainly part of the nucleus and involved in nucleosome assembly along with DNA binding. Additionally, highly abundant proteins in EVs of HF group were found to be involved in spermatogenesis, motility, acrosome reaction, capacitation, gamete fusion, and cryotolerance. Two highly abundant proteins, protein disulfide-isomerase A4 and gelsolin, are associated with sperm-oocyte fusion and acrosome reaction, respectively, and their immunolocalization on spermatozoa may indicate that these proteins are transferred through EVs. Our evidences support that proteins in EVs and subsequently their presence on sperm, are strongly associated with sperm functions. Altogether, our investigation indicates that SPEVs possess crucial protein repertoires that are essential for enhancing sperm fertilizing capacity.
Collapse
Affiliation(s)
- Shiva Badrhan
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Seema Karanwal
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Ankit Pal
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Jatinder Singh Chera
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Vitika Chauhan
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Aditya Patel
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| | - Mukesh Bhakat
- ICAR- Central Institute of Research on Goat, Mathura, Uttar Pradesh, India
| | - Tirtha K. Datta
- Central Institute for Research on Buffaloes, Hisar, Haryana, India
| | - Rakesh Kumar
- Animal Genomics Laboratory, Animal Biotechnology Division, National Dairy Research Institute, Karnal, India
| |
Collapse
|
30
|
Yadav A, Xuan Y, Sen CK, Ghatak S. Standardized Reporting of Research on Exosomes to Ensure Rigor and Reproducibility. Adv Wound Care (New Rochelle) 2024. [PMID: 38888007 DOI: 10.1089/wound.2024.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024] Open
Abstract
Significance: The study of extracellular vesicles (EVs), especially exosomes, has unlocked new avenues in understanding cellular communication and potential therapeutic applications. Recent Advances: Advancements in EV research have shown significant contributions from the International Society for Extracellular Vesicles (ISEV), in establishing methodological standards. The evolution of the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines from 2014 to 2023 reflects enhanced research rigor and reproducibility. The launch of EV-TRACK platform promotes uniformity and reproducibility by providing a centralized repository for data sharing and standardization practices. Furthermore, databases like EVpedia and ExoCarta have facilitated data sharing and collaboration within the scientific community. Concurrently, exosome-based therapies have emerged as a forefront area within regenerative medicine and targeted drug delivery, showcasing the potential of exosomes in promoting tissue regeneration. Critical Issues: Despite advancements, the field grapples with challenges such as vesicular heterogeneity, EV isolation complexity, and standardization. These issues impact research reproducibility and clinical applications. The inconsistency in exosomal preparations in clinical trials poses significant challenges to therapeutic efficacy and safety. Future Directions: The review outlines critical areas for future research, including the need for technological innovation in EV isolation and characterization, the establishment of standardized protocols, and a deeper understanding of exosome biology. The review also highlights the need to reassess guidelines, develop new EV isolation and characterization technologies, and establish standardized protocols to overcome current limitations. Emphasis is placed on interdisciplinary research and collaboration to address the complexities of EV biology, improve clinical trial design, and ultimately realize exosome's therapeutic and diagnostic potential. Continued evaluation and rigorous scientific validation are essential for successful exosome integration.
Collapse
Affiliation(s)
- Anita Yadav
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yi Xuan
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chandan K Sen
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Morganti C, Bonora M, Ito K. Metabolism and HSC fate: what NADPH is made for. Trends Cell Biol 2024:S0962-8924(24)00141-7. [PMID: 39054107 DOI: 10.1016/j.tcb.2024.07.003] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial metabolism plays a central role in the regulation of hematopoietic stem cell (HSC) biology. Mitochondrial fatty acid oxidation (FAO) is pivotal in controlling HSC self-renewal and differentiation. Herein, we discuss recent evidence suggesting that NADPH generated in the mitochondria can influence the fate of HSCs. Although NADPH has multiple functions, HSCs show high levels of NADPH that are preferentially used for cholesterol biosynthesis. Endogenous cholesterol supports the biogenesis of extracellular vesicles (EVs), which are essential for maintaining HSC properties. We also highlight the significance of EVs in hematopoiesis through autocrine signaling. Elucidating the mitochondrial NADPH-cholesterol axis as part of the metabolic requirements of healthy HSCs will facilitate the development of new therapies for hematological disorders.
Collapse
Affiliation(s)
- Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| |
Collapse
|
32
|
Kazeminava F, Javanbakht S, Latifi Z, Rasoulzadehzali M, Abbaszadeh M, Alimohammadzadeh B, Mahdipour M, Fattahi A, Hamishehkar H, Adibag Z, Nouri M. Ultrasound-assisted encapsulating folic acid-based carbon quantum dots within breast cancer cell-derived exosomes as a co-receptors-mediated anticancer nanocarrier for enhanced breast cancer therapy. Sci Rep 2024; 14:16941. [PMID: 39043763 PMCID: PMC11266556 DOI: 10.1038/s41598-024-67934-6] [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: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
The nonspecific nature of cancer drug delivery often results in substantial toxic side effects during treatments for breast cancer. To mitigate these negative outcomes, our approach involves loading methotrexate (MTX) within carbon quantum dots (CQDs) synthesized from folic acid, which are then enveloped in exosomal membranes obtained from breast cancer cells (Ex@MTX-CQDs). Analysis utilizing nanoparticle tracking techniques has demonstrated that these Ex@MTX-CQDs maintain the physical and biochemical properties of their exosomal precursors. The release profile of MTX indicated a restricted release percentage (less than 10%) under normal physiological conditions, which is contrasted by a more consistent release rate (approximately 65%) when emulating the conditions found within tumor tissues. The toxicological assessments have confirmed that the presence of exosomes combined with leftover folic acid significantly improves the delivery efficacy of MTX directly to the cancerous cells through the binding to folate and heparan sulfate proteoglycan receptors. This process results in increased disruption of the mitochondrial membrane potential and subsequently triggers apoptosis, ultimately leading to the destruction of cancerous cells. Our research could potentially contribute to the further innovation and application of nanocarriers derived from biological sources for the targeted treatment of breast cancer.
Collapse
Affiliation(s)
- Fahimeh Kazeminava
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siamak Javanbakht
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mahmoud Abbaszadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Adibag
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Gonçalves PP, da Silva CL, Bernardes N. Advancing cancer therapeutics: Integrating scalable 3D cancer models, extracellular vesicles, and omics for enhanced therapy efficacy. Adv Cancer Res 2024; 163:137-185. [PMID: 39271262 DOI: 10.1016/bs.acr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Cancer remains as one of the highest challenges to human health. However, anticancer drugs exhibit one of the highest attrition rates compared to other therapeutic interventions. In part, this can be attributed to a prevalent use of in vitro models with limited recapitulative potential of the in vivo settings. Three dimensional (3D) models, such as tumor spheroids and organoids, offer many research opportunities to address the urgent need in developing models capable to more accurately mimic cancer biology and drug resistance profiles. However, their wide adoption in high-throughput pre-clinical studies is dependent on scalable manufacturing to support large-scale therapeutic drug screenings and multi-omic approaches for their comprehensive cellular and molecular characterization. Extracellular vesicles (EVs), which have been emerging as promising drug delivery systems (DDS), stand to significantly benefit from such screenings conducted in realistic cancer models. Furthermore, the integration of these nanomedicines with 3D cancer models and omics profiling holds the potential to deepen our understanding of EV-mediated anticancer effects. In this chapter, we provide an overview of the existing 3D models used in cancer research, namely spheroids and organoids, the innovations in their scalable production and discuss how omics can facilitate the implementation of these models at different stages of drug testing. We also explore how EVs can advance drug delivery in cancer therapies and how the synergy between 3D cancer models and omics approaches can benefit in this process.
Collapse
Affiliation(s)
- Pedro P Gonçalves
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Nuno Bernardes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
34
|
Chen Y, Zhang Z, Li Z, Wu W, Lan S, Yan T, Mei K, Qiao Z, Wang C, Bai C, Li Z, Wu S, Wang J, Zhang Q. Dynamic nanomechanical characterization of cells in exosome therapy. MICROSYSTEMS & NANOENGINEERING 2024; 10:97. [PMID: 39015940 PMCID: PMC11251037 DOI: 10.1038/s41378-024-00735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/20/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been confirmed to enhance cell proliferation and improve tissue repair. Exosomes release their contents into the cytoplasmic solution of the recipient cell to mediate cell expression, which is the main pathway through which exosomes exert therapeutic effects. The corresponding process of exosome internalization mainly occurs in the early stage of treatment. However, the therapeutic effect of exosomes in the early stage remains to be further studied. We report that the three-dimensional cell traction force can intuitively reflect the ability of exosomes to enhance the cytoskeleton and cell contractility of recipient cells, serving as an effective method to characterize the therapeutic effect of exosomes. Compared with traditional biochemical methods, we can visualize the early therapeutic effect of exosomes in real time without damage by quantifying the cell traction force. Through quantitative analysis of traction forces, we found that endometrial stromal cells exhibit short-term cell roundness accompanied by greater traction force during the early stage of exosome therapy. Further experiments revealed that exosomes enhance the traction force and cytoskeleton by regulating the Rac1/RhoA signaling pathway, thereby promoting cell proliferation. This work provides an effective method for rapidly quantifying the therapeutic effects of exosomes and studying the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ye Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Ziwei Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Wenjie Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shihai Lan
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Tianhao Yan
- Department of Cell Biology and Genetics, College of Basic Medical Sciences, Jilin University, Changchun, 130021 China
| | - Kainan Mei
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Zihan Qiao
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chen Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Chuanbiao Bai
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Ziyan Li
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Shangquan Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Science, 15 Beisihuan West Road, Beijing, 100190 China
| | - Jianye Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, 230022 China
| | - Qingchuan Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027 China
| |
Collapse
|
35
|
Satheeshan G, Si AK, Rutta J, Venkatesh T. Exosome theranostics: Comparative analysis of P body and exosome proteins and their mutations for clinical applications. Funct Integr Genomics 2024; 24:124. [PMID: 38995459 DOI: 10.1007/s10142-024-01404-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024]
Abstract
Exosomes are lipid-bilayered vesicles, originating from early endosomes that capture cellular proteins and genetic materials to form multi-vesicular bodies. These exosomes are secreted into extracellular fluids such as cerebrospinal fluid, blood, urine, and cell culture supernatants. They play a key role in intercellular communication by carrying active molecules like lipids, cytokines, growth factors, metabolites, proteins, and RNAs. Recently, the potential of exosomal delivery for therapeutic purposes has been explored due to their low immunogenicity, nano-scale size, and ability to cross cellular barriers. This review comprehensively examines the biogenesis of exosomes, their isolation techniques, and their diverse applications in theranostics. We delve into the mechanisms and methods for loading exosomes with mRNA, miRNA, proteins, and drugs, highlighting their transformative role in delivering therapeutic payloads. Additionally, the utility of exosomes in stem cell therapy is discussed, showcasing their potential in regenerative medicine. Insights into exosome cargo using pre- or post-loading techniques are critical for exosome theranostics. We review exosome databases such as ExoCarta, Expedia, and ExoBCD, which document exosome cargo. From these databases, we identified 25 proteins common to both exosomes and P-bodies, known for mutations in the COSMIC database. Exosome databases do not integrate with mutation analysis programs; hence, we performed mutation analysis using additional databases. Accounting for the mutation status of parental cells and exosomal cargo is crucial in exosome theranostics. This review provides a comprehensive report on exosome databases, proteins common to exosomes and P-bodies, and their mutation analysis, along with the latest studies on exosome-engineered theranostics.
Collapse
Affiliation(s)
- Greeshma Satheeshan
- Dept of Biochemistry and Molecular Biology, Central University of Kerala, Krishna building, Periye, Kasargod, 671316, Kerala, India
| | - Ayan Kumar Si
- Dept of Biochemistry and Molecular Biology, Central University of Kerala, Krishna building, Periye, Kasargod, 671316, Kerala, India
| | - Joel Rutta
- Dept of Biochemistry and Molecular Biology, Central University of Kerala, Krishna building, Periye, Kasargod, 671316, Kerala, India
| | - Thejaswini Venkatesh
- Dept of Biochemistry and Molecular Biology, Central University of Kerala, Krishna building, Periye, Kasargod, 671316, Kerala, India.
| |
Collapse
|
36
|
Sun S, Li Q, Liu G, Huang X, Li A, Guo H, Qi L, Zhang J, Song J, Su X, Zhang Y. Endosomal protein DENND10/FAM45A integrates extracellular vesicle release with cancer cell migration. BMC Biol 2024; 22:154. [PMID: 38987765 PMCID: PMC11234546 DOI: 10.1186/s12915-024-01948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Affiliation(s)
- Shenqing Sun
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Qian Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ganggang Liu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaoheng Huang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Aiqing Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Haoran Guo
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Lijuan Qi
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jie Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jianrui Song
- Wisdom Lake Academy of Pharmacy, Jiangsu Provincial Higher Education Key Laboratory of Cell Therapy Nanoformulation, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| | - Xiong Su
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
- Suzhou Key Laboratory of Systems Biomedicine, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Yanling Zhang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
- Suzhou Key Laboratory of Systems Biomedicine, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
37
|
Samal S, Barik D, Shyamal S, Jena S, Panda AC, Dash M. Synergistic Interaction between Polysaccharide-Based Extracellular Matrix and Mineralized Osteoblast-Derived EVs Promotes Bone Regeneration via miRNA-mRNA Regulatory Axis. Biomacromolecules 2024; 25:4139-4155. [PMID: 38924768 DOI: 10.1021/acs.biomac.4c00269] [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: 06/28/2024]
Abstract
Extracellular vesicles (EVs) derived from bone progenitor cells are advantageous as cell-free and non-immunogenic cargo delivery vehicles. In this study, EVs are isolated from MC3T3-E1 cells before (GM-EVs) and after mineralization for 7 and 14 days (DM-EVs). It was observed that DM-EVs accelerate the process of differentiation in recipient cells more prominently. The small RNA sequencing of EVs revealed that miR-204-5p, miR-221-3p, and miR-148a-3p are among the highly upregulated miRNAs that have an inhibitory effect on the function of mRNAs, Sox11, Timp3, and Ccna2 in host cells, which is probably responsible for enhancing the activity of osteoblastic genes. To enhance the bioavailability of EVs, they are encapsulated in a chitosan-collagen composite hydrogel that serves as a bioresorbable extracellular matrix (ECM). The EVs-integrated scaffold (DM-EVs + Scaffold) enhances bone regeneration in critical-sized calvarial bone defects in rats within 8 weeks of implantation by providing the ECM cues. The shelf life of DM-EVs + Scaffold indicates that the bioactivity of EVs and their cargo in the polymer matrix remains intact for up to 30 days. Integrating mineralized cell-derived EVs into an ECM represents a bioresorbable matrix with a cell-free method for promoting new bone formation through the miRNA-mRNA regulatory axis.
Collapse
Affiliation(s)
- Sasmita Samal
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024 Odisha, India
| | - Debyashreeta Barik
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024 Odisha, India
| | - Sharmishtha Shyamal
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
- ICMR-National Institute for Reproduction Biology and Child Health, Mumbai 400012, India
| | - Sarita Jena
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| | - Amaresh C Panda
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| | - Mamoni Dash
- BRIC-Institute of Life Sciences (BRIC-ILS), Bhubaneswar 751023 Odisha, India
| |
Collapse
|
38
|
Peruzzi JA, Gunnels TF, Edelstein HI, Lu P, Baker D, Leonard JN, Kamat NP. Enhancing extracellular vesicle cargo loading and functional delivery by engineering protein-lipid interactions. Nat Commun 2024; 15:5618. [PMID: 38965227 PMCID: PMC11224323 DOI: 10.1038/s41467-024-49678-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: 04/29/2023] [Accepted: 06/13/2024] [Indexed: 07/06/2024] Open
Abstract
Naturally generated lipid nanoparticles termed extracellular vesicles (EVs) hold significant promise as engineerable therapeutic delivery vehicles. However, active loading of protein cargo into EVs in a manner that is useful for delivery remains a challenge. Here, we demonstrate that by rationally designing proteins to traffic to the plasma membrane and associate with lipid rafts, we can enhance loading of protein cargo into EVs for a set of structurally diverse transmembrane and peripheral membrane proteins. We then demonstrate the capacity of select lipid tags to mediate increased EV loading and functional delivery of an engineered transcription factor to modulate gene expression in target cells. We envision that this technology could be leveraged to develop new EV-based therapeutics that deliver a wide array of macromolecular cargo.
Collapse
Affiliation(s)
- Justin A Peruzzi
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Taylor F Gunnels
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Hailey I Edelstein
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Peilong Lu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA.
- Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, 60208, USA.
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA.
| | - Neha P Kamat
- Center for Synthetic Biology, Northwestern University, Evanston, IL, 60208, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA.
- Member, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, 60208, USA.
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
39
|
Slinning MS, Nthiga TM, Eichner C, Khadija S, Rome LH, Nilsen F, Dondrup M. Major vault protein is part of an extracellular cement material in the Atlantic salmon louse (Lepeophtheirus salmonis). Sci Rep 2024; 14:15240. [PMID: 38956386 PMCID: PMC11219742 DOI: 10.1038/s41598-024-65683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Major vault protein (MVP) is the main component of the vault complex, which is a highly conserved ribonucleoprotein complex found in most eukaryotic organisms. MVP or vaults have previously been found to be overexpressed in multidrug-resistant cancer cells and implicated in various cellular processes such as cell signaling and innate immunity. The precise function of MVP is, however, poorly understood and its expression and probable function in lower eukaryotes are not well characterized. In this study, we report that the Atlantic salmon louse expresses three full-length MVP paralogues (LsMVP1-3). Furthermore, we extended our search and identified MVP orthologues in several other ecdysozoan species. LsMVPs were shown to be expressed in various tissues at both transcript and protein levels. In addition, evidence for LsMVP to assemble into vaults was demonstrated by performing differential centrifugation. LsMVP was found to be highly expressed in cement, an extracellular material produced by a pair of cement glands in the adult female salmon louse. Cement is important for the formation of egg strings that serve as protective coats for developing embryos. Our results imply a possible novel function of LsMVP as a secretory cement protein. LsMVP may play a role in structural or reproductive functions, although this has to be further investigated.
Collapse
Affiliation(s)
- Malene Skuseth Slinning
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Thaddaeus Mutugi Nthiga
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Christiane Eichner
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Syeda Khadija
- Department of Biological Chemistry, David Geffen School of Medicine and the California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine and the California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Frank Nilsen
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Michael Dondrup
- SLRC, Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Pb. 7803, 5020, Bergen, Norway.
| |
Collapse
|
40
|
Kaur M, Fusco S, Van den Broek B, Aseervatham J, Rostami A, Iacovitti L, Grassi C, Lukomska B, Srivastava AK. Most recent advances and applications of extracellular vesicles in tackling neurological challenges. Med Res Rev 2024; 44:1923-1966. [PMID: 38500405 DOI: 10.1002/med.22035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Over the past few decades, there has been a notable increase in the global burden of central nervous system (CNS) diseases. Despite advances in technology and therapeutic options, neurological and neurodegenerative disorders persist as significant challenges in treatment and cure. Recently, there has been a remarkable surge of interest in extracellular vesicles (EVs) as pivotal mediators of intercellular communication. As carriers of molecular cargo, EVs demonstrate the ability to traverse the blood-brain barrier, enabling bidirectional communication. As a result, they have garnered attention as potential biomarkers and therapeutic agents, whether in their natural form or after being engineered for use in the CNS. This review article aims to provide a comprehensive introduction to EVs, encompassing various aspects such as their diverse isolation methods, characterization, handling, storage, and different routes for EV administration. Additionally, it underscores the recent advances in their potential applications in neurodegenerative disorder therapeutics. By exploring their unique capabilities, this study sheds light on the promising future of EVs in clinical research. It considers the inherent challenges and limitations of these emerging applications while incorporating the most recent updates in the field.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bram Van den Broek
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jaya Aseervatham
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Jefferson Stem Cell and Regenerative Neuroscience Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Amit K Srivastava
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Liu Y, Lu S, Yang J, Yang Y, Jiao L, Hu J, Li Y, Yang F, Pang Y, Zhao Y, Gao Y, Liu W, Shu P, Ge W, He Z, Peng X. Analysis of the aging-related biomarker in a nonhuman primate model using multilayer omics. BMC Genomics 2024; 25:639. [PMID: 38926642 PMCID: PMC11209966 DOI: 10.1186/s12864-024-10556-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: 02/09/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Aging is a prominent risk factor for diverse diseases; therefore, an in-depth understanding of its physiological mechanisms is required. Nonhuman primates, which share the closest genetic relationship with humans, serve as an ideal model for exploring the complex aging process. However, the potential of the nonhuman primate animal model in the screening of human aging markers is still not fully exploited. Multiomics analysis of nonhuman primate peripheral blood offers a promising approach to evaluate new therapies and biomarkers. This study explores aging-related biomarker through multilayer omics, including transcriptomics (mRNA, lncRNA, and circRNA) and proteomics (serum and serum-derived exosomes) in rhesus monkeys (Macaca mulatta). RESULTS Our findings reveal that, unlike mRNAs and circRNAs, highly expressed lncRNAs are abundant during the key aging period and are associated with cancer pathways. Comparative analysis highlighted exosomal proteins contain more types of proteins than serum proteins, indicating that serum-derived exosomes primarily regulate aging through metabolic pathways. Finally, eight candidate aging biomarkers were identified, which may serve as blood-based indicators for detecting age-related brain changes. CONCLUSIONS Our results provide a comprehensive understanding of nonhuman primate blood transcriptomes and proteomes, offering novel insights into the aging mechanisms for preventing or treating age-related diseases.
Collapse
Affiliation(s)
- Yunpeng Liu
- State Key Laboratory of Respiratory Health and Multimorbidity, National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, CAMS & PUMC, Beijing, 100021, China
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Jing Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Li Jiao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Jingwen Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yunli Pang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China
| | - Yanpan Gao
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Wei Ge
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China.
| | - Xiaozhong Peng
- State Key Laboratory of Respiratory Health and Multimorbidity, National Center of Technology Innovation for Animal Model, National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Sciences, CAMS & PUMC, Beijing, 100021, China.
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, 650031, China.
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, CAMS & PUMC, Beijing, 100005, China.
| |
Collapse
|
42
|
Patel SK, Bons J, Rose JP, Chappel JR, Beres RL, Watson MA, Webster C, Burton JB, Bruderer R, Desprez PY, Reiter L, Campisi J, Baker ES, Schilling B. Exosomes Released from Senescent Cells and Circulatory Exosomes Isolated from Human Plasma Reveal Aging-associated Proteomic and Lipid Signatures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.22.600215. [PMID: 38979258 PMCID: PMC11230204 DOI: 10.1101/2024.06.22.600215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Senescence emerged as a significant mechanism of aging and age-related diseases, offering an attractive target for clinical interventions. Senescent cells release a senescence-associated secretory phenotype (SASP), including exosomes that may act as signal transducers between distal tissues, propagating secondary or bystander senescence and signaling throughout the body. However, the composition of exosome SASP remains underexplored, presenting an opportunity for novel unbiased discovery. Here, we present a detailed proteomic and lipidomic analysis of exosome SASP using mass spectrometry from human plasma from young and older individuals and from tissue culture of senescent primary human lung fibroblasts. We identified ~1,300 exosome proteins released by senescent fibroblasts induced by three different senescence inducers causing most exosome proteins to be differentially regulated with senescence. In parallel, a human plasma cohort from young and old individuals revealed over 1,350 exosome proteins and 171 plasma exosome proteins were regulated when comparing old vs young individuals. Of the age-regulated plasma exosome proteins, we observed 52 exosome SASP factors that were also regulated in exosomes from the senescent fibroblasts, including serine protease inhibitors (SERPINs), Prothrombin, Coagulation factor V, Plasminogen, and Reelin. In addition, 247 lipids were identified with high confidence in all exosome samples. Following the senescence inducers, a majority of the identified phosphatidylcholine, phosphatidylethanolamine, and sphingomyelin species increased significantly indicating cellular membrane changes. The most notable categories of significantly changed proteins were related to extracellular matrix remodeling and inflammation, both potentially detrimental pathways that can damage surrounding tissues and even induce secondary or bystander senescence. Our findings reveal mechanistic insights and potential senescence biomarkers, enabling a better approach to surveilling the senescence burden in the aging population and offering promising therapeutic targets for interventions.
Collapse
|
43
|
Wang Z, Zhang Y, Li X. Mitigation of Oxidative Stress in Idiopathic Pulmonary Fibrosis Through Exosome-Mediated Therapies. Int J Nanomedicine 2024; 19:6161-6176. [PMID: 38911503 PMCID: PMC11193999 DOI: 10.2147/ijn.s453739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/01/2024] [Indexed: 06/25/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) poses a formidable clinical challenge, characterized by the thickening of alveolar septa and the onset of pulmonary fibrosis. The pronounced activation of oxidative stress emerges as a pivotal hallmark of inflammation. Traditional application of exogenous antioxidants proves inadequate in addressing oxidative stress, necessitating exploration into strategies to augment their antioxidant efficacy. Exosomes, nano-sized extracellular vesicles harboring a diverse array of bioactive factors, present as promising carriers with the potential to meet this challenge. Recent attention has been directed towards the clinical applications of exosomes in IPF, fueling the impetus for this comprehensive review. We have compiled fresh insights into the role of exosomes in modulating oxidative stress in IPF and delved into their potential as carriers for regulating endogenous reactive oxygen species generation. This review endeavors to bridge the divide between exosome research and IPF, traversing from bedside to bench. Through the synthesis of recent findings, we propose exosomes as a novel and promising strategy for improving the outcomes of IPF therapy.
Collapse
Affiliation(s)
- Zaiyan Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, People’s Republic of China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| | - Xiaoning Li
- Department of Geriatric Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, People’s Republic of China
| |
Collapse
|
44
|
Bhavsar SP, Morini M. The emerging role of the exosomal proteins in neuroblastoma. Front Oncol 2024; 14:1414063. [PMID: 38962276 PMCID: PMC11219817 DOI: 10.3389/fonc.2024.1414063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Exosomes are a subclass of extracellular vesicles shown to promote the cancer growth and support metastatic progression. The proteomic analysis of neuroblastoma-derived exosomes has revealed proteins involved in cell migration, proliferation, metastasis, and in the modulation of tumor microenvironment - thus contributing to the tumor development and an aggressive metastatic phenotype. This review gives an overview of the current understanding of the exosomal proteins in neuroblastoma and of their potential as diagnostic/prognostic biomarker of disease and therapeutics.
Collapse
Affiliation(s)
- Swapnil Parashram Bhavsar
- Pediatric Research Group, Department of Clinical Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Martina Morini
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
45
|
Sani F, Shojaei S, Tabatabaei SA, Khorraminejad-Shirazi M, Latifi M, Sani M, Azarpira N. CAR-T cell-derived exosomes: a new perspective for cancer therapy. Stem Cell Res Ther 2024; 15:174. [PMID: 38886844 PMCID: PMC11184895 DOI: 10.1186/s13287-024-03783-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell adoptive immunotherapy is a promising cancer treatment that uses genetically engineered T cells to attack tumors. However, this therapy can have some adverse effects. CAR-T cell-derived exosomes are a potential alternative to CAR-T cells that may overcome some limitations. Exosomes are small vesicles released by cells and can carry a variety of molecules, including proteins, RNA, and DNA. They play an important role in intercellular communication and can be used to deliver therapeutic agents to cancer cells. The application of CAR-T cell-derived exosomes could make CAR-T cell therapy more clinically controllable and effective. Exosomes are cell-free, which means that they are less likely to cause adverse reactions than CAR-T cells. The combination of CAR-T cells and exosomes may be a more effective way to treat cancer than either therapy alone. Exosomes can deliver therapeutic agents to cancer cells where CAR-T cells cannot reach. The appropriate application of both cellular and exosomal platforms could make CAR-T cell therapy a more practicable treatment for cancer. This combination therapy could offer a safe and effective way to treat a variety of cancers.
Collapse
Affiliation(s)
- Farnaz Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shabnam Shojaei
- School of Medicine, Shiraz Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammadhossein Khorraminejad-Shirazi
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Mona Latifi
- Department of Physiological Science, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Mahsa Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Negar Azarpira
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili Street, P.O. Box: 7193711351, Shiraz, Iran.
| |
Collapse
|
46
|
Wang K, Yang Z, Zhang B, Gong S, Wu Y. Adipose-Derived Stem Cell Exosomes Facilitate Diabetic Wound Healing: Mechanisms and Potential Applications. Int J Nanomedicine 2024; 19:6015-6033. [PMID: 38911504 PMCID: PMC11192296 DOI: 10.2147/ijn.s466034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/08/2024] [Indexed: 06/25/2024] Open
Abstract
Wound healing in diabetic patients is frequently hampered. Adipose-derived stem cell exosomes (ADSC-eoxs), serving as a crucial mode of intercellular communication, exhibit promising therapeutic roles in facilitating wound healing. This review aims to comprehensively outline the molecular mechanisms through which ADSC-eoxs enhance diabetic wound healing. We emphasize the biologically active molecules released by these exosomes and their involvement in signaling pathways associated with inflammation modulation, cellular proliferation, vascular neogenesis, and other pertinent processes. Additionally, the clinical application prospects of the reported ADSC-eoxs are also deliberated. A thorough understanding of these molecular mechanisms and potential applications is anticipated to furnish a theoretical groundwork for combating diabetic wound healing.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zihui Yang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Boyu Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
47
|
Lavi Arab F, Hoseinzadeh A, Hafezi F, Sadat Mohammadi F, Zeynali F, Hadad Tehran M, Rostami A. Mesenchymal stem cell-derived exosomes for management of prostate cancer: An updated view. Int Immunopharmacol 2024; 134:112171. [PMID: 38701539 DOI: 10.1016/j.intimp.2024.112171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024]
Abstract
Prostate cancer represents the second most prevalent form of cancer found in males, and stands as the fifth primary contributor to cancer-induced mortality on a global scale. Research has shown that transplanted mesenchymal stem cells (MSCs) can migrate by homing to tumor sites in the body. In prostate cancer, researchers have explored the fact that MSC-based therapies (including genetically modified delivery vehicles or vectors) and MSC-derived exosomes are emerging as attractive options to improve the efficacy and safety of traditional cancer therapies. In addition, researchers have reported new insights into the application of extracellular vesicle (EV)-MSC therapy as a novel treatment option that could provide a more effective and targeted approach to prostate cancer treatment. Moreover, the new generation of exosomes, which contain biologically functional molecules as signal transducers between cells, can simultaneously deliver different therapeutic agents and induce an anti-tumor phenotype in immune cells and their recruitment to the tumor site. The results of the current research on the use of MSCs in the treatment of prostate cancer may be helpful to researchers and clinicians working in this field. Nevertheless, it is crucial to emphasize that although dual-role MSCs show promise as a therapeutic modality for managing prostate cancer, further investigation is imperative to comprehensively grasp their safety and effectiveness. Ongoing clinical trials are being conducted to assess the viability of MSCs in the management of prostate cancer. The results of these trials will help determine the viability of this approach. Based on the current literature, engineered MSCs-EV offer great potential for application in targeted tumor therapy.
Collapse
Affiliation(s)
- Fahimeh Lavi Arab
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.; Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Hafezi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Sadat Mohammadi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farid Zeynali
- Department of Urology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Melika Hadad Tehran
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amirreza Rostami
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
48
|
Janković T, Janković M. Extracellular vesicles and glycans: new avenue for biomarker research. Biochem Med (Zagreb) 2024; 34:020503. [PMID: 38882582 PMCID: PMC11177654 DOI: 10.11613/bm.2024.020503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Abstract
The investigation of biomarkers is constantly evolving. New molecules and molecular assemblies, such as soluble and particulate complexes, emerged as biomarkers from basic research and investigation of different proteomes, genomes, and glycomes. Extracellular vesicles (EVs), and glycans, complex carbohydrates are ubiquitous in nature. The composition and structure of both reflect physiological state of paternal cells and are strikingly changed in diseases. The EV-associated glycans, alone or in combination with soluble glycans in related biological fluids, used as analytes, aim to capture full complex biomarker picture, enabling its use in different clinical settings. Bringing together EVs and glycans can help to extract meaningful data from their extreme and distinct heterogeneities for use in the real-time diagnostics. The glycans on the surface of EVs could mark their subpopulations and establish the glycosignature, the solubilisation signature and molecular patterns. They all contribute to a new way of looking at and looking for composite biomarkers.
Collapse
Affiliation(s)
- Tamara Janković
- Department for Immunochemistry and Glycobiology, Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Belgrade, Serbia
| | - Miroslava Janković
- Department for Immunochemistry and Glycobiology, Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
49
|
Khoo A, Govindarajan M, Qiu Z, Liu LY, Ignatchenko V, Waas M, Macklin A, Keszei A, Neu S, Main BP, Yang L, Lance RS, Downes MR, Semmes OJ, Vesprini D, Liu SK, Nyalwidhe JO, Boutros PC, Kislinger T. Prostate cancer reshapes the secreted and extracellular vesicle urinary proteomes. Nat Commun 2024; 15:5069. [PMID: 38871730 PMCID: PMC11176296 DOI: 10.1038/s41467-024-49424-5] [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: 08/24/2023] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Urine is a complex biofluid that reflects both overall physiologic state and the state of the genitourinary tissues through which it passes. It contains both secreted proteins and proteins encapsulated in tissue-derived extracellular vesicles (EVs). To understand the population variability and clinical utility of urine, we quantified the secreted and EV proteomes from 190 men, including a subset with prostate cancer. We demonstrate that a simple protocol enriches prostatic proteins in urine. Secreted and EV proteins arise from different subcellular compartments. Urinary EVs are faithful surrogates of tissue proteomes, but secreted proteins in urine or cell line EVs are not. The urinary proteome is longitudinally stable over several years. It can accurately and non-invasively distinguish malignant from benign prostatic lesions and can risk-stratify prostate tumors. This resource quantifies the complexity of the urinary proteome and reveals the synergistic value of secreted and EV proteomes for translational and biomarker studies.
Collapse
Affiliation(s)
- Amanda Khoo
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Meinusha Govindarajan
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Zhuyu Qiu
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Urology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Lydia Y Liu
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90024, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Urology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Matthew Waas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Andrew Macklin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Alexander Keszei
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada
| | - Sarah Neu
- Division of Surgery, Urology, Sunnybrook Health Sciences Centre, Toronto, ON, M4N 3M5, Canada
| | - Brian P Main
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | | | - Michelle R Downes
- Division of Anatomic Pathology, Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON, M4N 3M5, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - O John Semmes
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Danny Vesprini
- Department of Radiation Oncology, University of Toronto, Toronto, ON, M5T 1P5, Canada
- Odette Cancer Research Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Stanley K Liu
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, M5T 1P5, Canada
- Odette Cancer Research Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Julius O Nyalwidhe
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Paul C Boutros
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90024, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Urology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, 90095, USA.
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2C1, Canada.
| |
Collapse
|
50
|
Dogny C, André-Lévigne D, Kalbermatten DF, Madduri S. Therapeutic Potential and Challenges of Mesenchymal Stem Cell-Derived Exosomes for Peripheral Nerve Regeneration: A Systematic Review. Int J Mol Sci 2024; 25:6489. [PMID: 38928194 PMCID: PMC11203969 DOI: 10.3390/ijms25126489] [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: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Gap injuries to the peripheral nervous system result in pain and loss of function, without any particularly effective therapeutic options. Within this context, mesenchymal stem cell (MSC)-derived exosomes have emerged as a potential therapeutic option. Thus, the focus of this study was to review currently available data on MSC-derived exosome-mounted scaffolds in peripheral nerve regeneration in order to identify the most promising scaffolds and exosome sources currently in the field of peripheral nerve regeneration. We conducted a systematic review following PRISMA 2020 guidelines. Exosome origins varied (adipose-derived MSCs, bone marrow MSCs, gingival MSC, induced pluripotent stem cells and a purified exosome product) similarly to the materials (Matrigel, alginate and silicone, acellular nerve graft [ANG], chitosan, chitin, hydrogel and fibrin glue). The compound muscle action potential (CMAP), sciatic functional index (SFI), gastrocnemius wet weight and histological analyses were used as main outcome measures. Overall, exosome-mounted scaffolds showed better regeneration than scaffolds alone. Functionally, both exosome-enriched chitin and ANG showed a significant improvement over time in the sciatica functional index, CMAP and wet weight. The best histological outcomes were found in the exosome-enriched ANG scaffold with a high increase in the axonal diameter and muscle cross-section area. Further studies are needed to confirm the efficacy of exosome-mounted scaffolds in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Clelia Dogny
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Dominik André-Lévigne
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Daniel F. Kalbermatten
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland
| | - Srinivas Madduri
- Department of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|