1
|
Sahu VK, Lokhande KB, Swamy VK, Basu S, Ranjan A. Role of the transmembrane polar residues on CD151 in cholesterol binding. J Biomol Struct Dyn 2025:1-13. [PMID: 39743780 DOI: 10.1080/07391102.2024.2446663] [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: 04/25/2024] [Accepted: 07/30/2024] [Indexed: 01/04/2025]
Abstract
Tetraspanins superfamily proteins have been shown to play an important role in several physiological processes and diseases such as cancer. Transmembrane polar residues of tetraspanins have an implication in regulating the process of cancer metastasis. Tetraspanin CD82 has been demonstrated to exert an anti-metastatic role while mutating polar residues in its transmembrane domains (TMDs) abrogates its metastasis inhibitory role. However, CD151, being pro-metastatic tetraspanin, the role of polar residues in TMDs of CD151 in cholesterol binding and its stability has not been investigated. In this study, we employed bioinformatics analysis of CD151 sequences to determine polar residue in TMDs, molecular docking, dynamics, and normal mode analysis studies to investigate the contribution of polar residues on cholesterol binding by modelling and comparing wild type with polar residue mutant of CD151. The study finds three polar residues viz Asn24, Glu104 and Gln234, in the TMD1, TMD2, and TMD4, respectively, in CD151 that are conserved in most vertebrates indicating their functional significance. Furthermore, we found that these polar residues are required for CD151 stability and facilitate stable interactions with cholesterol. Further cholesterol binding influenced the differences in the distance between large and small extracellular domains of CD151 indicating its important role in the open and closed conformation of the protein. In summary, CD151 exists in open and closed conformation by regulating its interactions with cholesterol. This interaction may modulate the cell membrane dynamics suggesting its potential implication in cancer metastasis.
Collapse
Affiliation(s)
- Vishal Kumar Sahu
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Patil Vidyapeeth, Pune, Maharashtra, India
- Translational Bioinformatics and Computational Genomics Research Lab, Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Venkateswar K Swamy
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Patil Vidyapeeth, Pune, Maharashtra, India
- Bioinformatics Research Group, MIT School of Bioengineering Science and Research, MIT- ADT University, Pune, Maharashtra, India
| | - Soumya Basu
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Amit Ranjan
- Cancer and Translational Research Centre, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
2
|
An J, Chen B, Zhang R, Tian D, Shi K, Zhang L, Zhang G, Wang J, Yang H. Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes in Spinal Cord Injury. Mol Neurobiol 2025; 62:1291-1315. [PMID: 39312070 DOI: 10.1007/s12035-024-04490-0] [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: 09/11/2023] [Accepted: 09/10/2024] [Indexed: 01/04/2025]
Abstract
Spinal cord injury (SCI) can lead to severe motor and sensory dysfunction, with a high rate of disability and mortality. Due to the complicated pathological process of SCI, there is no effective clinical treatment strategy at present. Although mesenchymal stem cells (MSCs) are effective in the treatment of SCI, their application is limited by factors such as low survival rate, cell dedifferentiation, tumorigenesis, blood-brain barrier, and immune rejection. Fortunately, there is growing evidence that most of the biological and therapeutic effects of MSCs may be mediated by the release of paracrine factors, which are extracellular vesicles called exosomes. Exosomes are small endosomal vesicles with bilaminar membranes that have recently been recognized as key mediators for communication between cells and tissues through the transfer of proteins, lipids, nucleic acids, cytokines, and growth factors. Mesenchymal stem cell-derived exosomes (MSC-exos) play a critical role in SCI repair by promoting angiogenesis and axonal growth, regulating inflammation and immune response, inhibiting apoptosis, and maintaining the integrity of the blood-spinal cord barrier. Furthermore, they can be used to transport genetic material or drugs to target cells, and their relatively small size allows them to permeate the blood-brain barrier. Studies have demonstrated that some exosomal miRNAs derived from MSCs play a significant role in the treatment of SCI. In this review, we summarize recent research advances in MSC-exos and exosomal miRNAs in SCI therapy to better understand this emerging cell-free therapeutic strategy and discuss the advantages and challenges of MSC-exos in future clinical applications.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Bo Chen
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China.
| | - Rui Zhang
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, 550081, Guizhou, China
| | - Ding Tian
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Kuohao Shi
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Lingling Zhang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Gaorong Zhang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Jingchao Wang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China
| | - Hao Yang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Beilin District, 555 East Youyi Road, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
3
|
Xia X, Shen P, Yang G, Yao M, Wu X, Lyu L, He Y, Li Z, Wang W, Yang Y, Ao X, Xia C, Chen Z, Xu X. The proliferation/migration ability mediated by CD151/PI3K/AKT pathway determines the therapeutic effect of hUC-MSCs transplantation on rheumatoid arthritis. Clin Exp Hypertens 2024; 46:2366270. [PMID: 38864268 DOI: 10.1080/10641963.2024.2366270] [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: 02/14/2024] [Accepted: 03/04/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVE To elucidate the underlying mechanism by which the proliferation and migration abilities of human umbilical cord mesenchymal stem cells (hUC-MSCs) determine their therapeutic efficacy in rheumatoid arthritis treatment. METHODS The DBA/1J mice were utilized to establish a collagen-induced RA (CIA) mouse model and to validate the therapeutic efficacy of hUC-MSCs transfected with CD151 siRNA. RNA-seq, QT-PCR and western blotting were utilized to evaluate the mRNA and protein levels of the PI3K/AKT pathway, respectively. RESULTS IFN-γ significantly enhanced the proliferation and migration abilities of hUC-MSCs, up-regulating the expression of CD151, a gene related to cell proliferation and migration. Effective inhibition of this effect was achieved through CD151 siRNA treatment. However, IFN-γ did not affect hUC-MSCs differentiation or changes in cell surface markers. Additionally, transplantation of CD151-interfered hUC-MSCs (siRNA-CD151-hUC-MSCs) resulted in decreased colonization in the toes of CIA mice and worse therapeutic effects compared to empty vector treatment (siRNA-NC-hUC-MSCs). CONCLUSION IFN-γ facilitates the proliferation and migration of hUC-MSCs through the CD151/PI3K/AKT pathway. The therapeutic efficacy of siRNA-CD151-hUC-MSCs was found to be inferior to that of siRNA-NC-hUC-MSCs.
Collapse
Affiliation(s)
- Xuewei Xia
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of senile Cardio-cerebrovascular diseases, Ministry of Education, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Peixin Shen
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Guomei Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Mengwei Yao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaofeng Wu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Lina Lyu
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of senile Cardio-cerebrovascular diseases, Ministry of Education, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yanji He
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of senile Cardio-cerebrovascular diseases, Ministry of Education, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Zhuxin Li
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of senile Cardio-cerebrovascular diseases, Ministry of Education, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Wei Wang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
- Key Laboratory of senile Cardio-cerebrovascular diseases, Ministry of Education, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, China
| | - Yi Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Chuanjiang Xia
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhuo Chen
- Department of General Surgery, The 906th Hospital of PLA, Ningbo, Zhejiang, China
| | - Xiang Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Stem Cell and Regenerative Medicine, Daping Hospital, Army Medical University, Chongqing, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Najdaghi S, Davani DN, Fouladseresht H, Ebrahimi N, Sullman MJM, Moradi M, Eskandari N. The Role of Extracellular Vesicles and Microparticles in Central Nervous System Disorders: Mechanisms, Biomarkers, and Therapeutic Potential. Cell Mol Neurobiol 2024; 44:82. [PMID: 39625540 PMCID: PMC11614997 DOI: 10.1007/s10571-024-01518-w] [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: 07/27/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Microscopic, membranous vesicles known as extracellular vesicles (EVs) have been proposed to play a role in the mechanisms underlying central nervous system (CNS) diseases. EVs are secreted by a variety of cells, including myeloid, endothelial, microglial, oligodendroglial, and mesenchymal stem cells (MSCs). Body fluids such as plasma, urine, and cerebrospinal fluid (CSF) contain microparticles (MPs). The detection of MPs in CSF may indicate genetic or environmental susceptibility to conditions such as schizophrenia, schizoaffective disorder, and bipolar disorder. MPs of different origins can exhibit changes in specific biomarkers at various stages of the disease, aiding in the diagnosis and monitoring of neurological conditions. However, understanding the role and clinical applications of MPs is complicated by challenges such as their isolation and dual roles within the CNS. In this review, we discuss the history, characteristics, and roles of MPs in CNS diseases. We also provide practical insights for future research and highlight the challenges that obscure the therapeutic potential of MPs.
Collapse
Affiliation(s)
- Soroush Najdaghi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hamed Fouladseresht
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Ebrahimi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Social Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Marjan Moradi
- Departement of Genetics, School of Science, Shahrekord University, Shahrakord, Iran
| | - Nahid Eskandari
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran.
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
Jung H, Jung Y, Seo J, Bae Y, Kim HS, Jeong W. Roles of extracellular vesicles from mesenchymal stem cells in regeneration. Mol Cells 2024; 47:100151. [PMID: 39547584 DOI: 10.1016/j.mocell.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are highly valued in regenerative medicine due to their ability to self-renew and differentiate into various cell types. Their therapeutic benefits are primarily due to their paracrine effects, in particular through extracellular vesicles (EVs), which are related to intercellular communication. Recent advances in EV production and extraction technologies highlight the potential of MSC-derived EVs (MSC-EVs) in tissue engineering and regenerative medicine. MSC-EVs offer several advantages over traditional cell therapies, including reduced toxicity and immunogenicity compared with whole MSCs. EVs carrying functional molecules such as growth factors, cytokines, and miRNAs play beneficial roles in tissue repair, fibrosis treatment, and scar prevention by promoting angiogenesis, skin cell migration, proliferation, extracellular matrix remodeling, and reducing inflammation. Despite the potential of MSC-EVs, there are several limitations to their use, including variability in quality, the need for standardized methods, low yield, and concerns about the composition of EVs and the potential risks. Overall, MSC-EVs are a promising alternative to cell-based therapies, and ongoing studies aim to understand their actions and optimize their use for better clinical outcomes in wound healing and skin regeneration.
Collapse
Affiliation(s)
- Hyeseong Jung
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yuyeon Jung
- Department of Dental Hygiene, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Junsik Seo
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Yeongju Bae
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Wooyoung Jeong
- Department of Biomedical Science, Catholic Kwandong University, Gangneung 25601, Republic of Korea; Research Center for Marine Bio-Food and Medicine, Catholic Kwandong University, Gangneung 25601, Republic of Korea.
| |
Collapse
|
6
|
Teng M, Luo J, Zhang Y, Reddy VRAP, Samuel P, Yao Y, Nair V. Viral miRNA delivered by exosomes from Marek's disease virus-transformed lymphoma cell line exerts regulatory function in internalized primary chicken embryo fibroblast cells. Tumour Virus Res 2024; 18:200286. [PMID: 38914377 PMCID: PMC11260597 DOI: 10.1016/j.tvr.2024.200286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/26/2024] Open
Abstract
In the past decade, research has demonstrated that viral miRNAs encoded by a number of viral genomes, particularly by most of the herpesvirus including Marek's disease virus (MDV), play important regulatory roles in viral infection, replication, and regulation of tumorigenesis. As macrovesicles in cells, exosomes can deliver viral miRNAs and exert gene regulatory functions. Whether the exosomes play a role in the replication, pathogenesis/tumorigenesis of avian herpesviruses such as oncogenic Marek's disease virus (MDV) remains unclear. Herein we extracted and identified the exosomes from MDV-transformed T cell line MSB-1 and demonstrated high abundance of MDV-1 miRNA expression. Using dual luciferase-based reporter assay, we also demonstrated that the exosomes derived from MSB-1 can deliver functional miRNA successfully into primary chicken embryo fibroblasts. These findings provide new insights into the role of exosomes and the mechanisms of how virus-encoded miRNA function in MDV latency/activation switching, viral replication, pathogenesis and/or tumorigenesis.
Collapse
Affiliation(s)
- Man Teng
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom; Institute for Animal Health & UK-China Centre of Excellence for Research on Avian Diseases, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Jun Luo
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom; Institute for Animal Health & UK-China Centre of Excellence for Research on Avian Diseases, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, People's Republic of China; Key Laboratory of Animal Immunology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450002, People's Republic of China
| | - Yaoyao Zhang
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom
| | - Vishwanatha R A P Reddy
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom
| | - Priya Samuel
- Department of Biological and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom.
| | - Venugopal Nair
- The Pirbright Institute & UK-China Centre of Excellence for Research on Avian Diseases, Pirbright, Guildford, Surrey, United Kingdom; Department of Biology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
7
|
Mizuta R, Kanao E, Ukyo K, Kuwada S, Sawada SI, Ishihama Y, Akiyoshi K, Sasaki Y. A Direct Approach for Living Biomembrane Printing on a Nanoparticle. NANO LETTERS 2024. [PMID: 39377259 DOI: 10.1021/acs.nanolett.4c03293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Biomembrane coating technologies have been developed to equip synthetic nanomaterials with natural biointerfaces. We report a one-step method for nondestructively coating the biomembranes of "living" cells onto nanoparticle surfaces. By using simple centrifugation, nanoparticles pass through a concentrated layer of living cells. This process mimics exosome release via endocytosis and exocytosis, preserving the membrane integrity of the source cells. The resulting silica nanoparticles were efficiently coated with membrane components from Raw264.7 cells. Nanoflow-liquid chromatography-tandem mass spectrometry confirmed that the proteins composing the membrane originated from the source cells. Additionally, the biomembrane coating suppressed the phagocytosis of silica nanoparticles by Raw264.7 cells while enhancing their uptake by HeLa cells. Our simple and efficient method for living biomembrane coating holds promise for the development of nanoparticles for medical and pharmaceutical applications.
Collapse
Affiliation(s)
- Ryosuke Mizuta
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Eisuke Kanao
- Laboratory of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Clinical and Analytical Chemistry, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Keigo Ukyo
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shusuke Kuwada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yasushi Ishihama
- Laboratory of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Clinical and Analytical Chemistry, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
8
|
Bhavsar V, Sahu A, Taware R. Stress-induced extracellular vesicles: insight into their altered proteomic composition and probable physiological role in cancer. Mol Cell Biochem 2024:10.1007/s11010-024-05121-x. [PMID: 39302488 DOI: 10.1007/s11010-024-05121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
EVs (extracellular vesicles) are phospholipid bilayer vesicles that can be released by both prokaryotic and eukaryotic cells in normal as well as altered physiological conditions. These vesicles also termed as signalosomes, possess a distinctive cargo comprising nucleic acids, proteins, lipids, and metabolites, enabling them to play a pivotal role in both local and long-distance intercellular communication. The composition, origin, and release of EVs can be influenced by different physiological conditions and a variety of stress factors, consequently affecting the contents carried within these vesicles. Therefore, identifying the modified contents of EVs can provide valuable insights into their functional role in stress-triggered communication. Particularly, this is important when EVs released from tumor microenvironment are investigated for their role in the development and dissemination of cancer. This review article emphasizes the importance of differential EV shedding and altered proteomic content in response to reduced oxygen concentration, altered levels of glucose and glutamine, pH variations, oxidative stress and Ca2+ ion concertation and it is subsequent effects on the behavior of recipient cells.
Collapse
Affiliation(s)
- Vaidehi Bhavsar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Ashish Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Ravindra Taware
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
9
|
Li L, Yao Z, Salimian KJ, Kong J, Zaheer A, Parian A, Gearhart SL, Mao HQ, Selaru FM. Extracellular Vesicles Delivered by a Nanofiber-Hydrogel Composite Enhance Healing In Vivo in a Model of Crohn's Disease Perianal Fistula. Adv Healthc Mater 2024:e2402292. [PMID: 39240055 DOI: 10.1002/adhm.202402292] [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: 06/22/2024] [Revised: 08/24/2024] [Indexed: 09/07/2024]
Abstract
Perianal fistulas represent a common, aggressive, and disabling complication of Crohn's disease (CD). Despite recent drug developments, novel surgical interventions as well as multidisciplinary treatment approaches, the outcome is dismal, with >50% therapy failure rates. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) offer potential therapeutic benefits for treating fistulizing CD, due to the pro-regenerative paracrine signals. However, a significant obstacle to clinical translation of EV-based therapy is the rapid clearance and short half-life of EVs in vivo. Here, an injectable, biodegradable nanofiber-hydrogel composite (NHC) microgel matrix that serves as a carrier to deliver MSC-derived EVs to a rat model of CD perianal fistula (PAF) is reported. It is found that EV-loaded NHC (EV-NHC) yields the best fistula healing when compared to other treatment arms. The MRI assessment reveals that the EV-NHC reduces inflammation at the fistula site and promotes tissue healing. The enhanced therapeutic outcomes are contributed by extended local retention and sustained release of EVs by NHC. In addition, the EV-NHC effectively reduces inflammation at the fistula site and promotes tissue healing and regeneration via macrophage polarization and neo-vascularization. This EV-NHC platform provides an off-the-shelf solution that facilitates its clinical translation.
Collapse
Affiliation(s)
- Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zhicheng Yao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevan J Salimian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiayuan Kong
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atif Zaheer
- Department of Radiology & Radiological Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Alyssa Parian
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Susan L Gearhart
- Division of Colorectal Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Florin M Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
10
|
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
|
11
|
Marquez J, Dong J, Hayashi J, Serrero G. Prostaglandin F2 Receptor Negative Regulator (PTGFRN) Expression Correlates With a Metastatic-like Phenotype in Epidermoid Carcinoma, Pediatric Medulloblastoma, and Mesothelioma. J Cell Biochem 2024; 125:e30616. [PMID: 38924562 DOI: 10.1002/jcb.30616] [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: 12/24/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024]
Abstract
Prostaglandin F2 receptor negative regulator (PTGFRN) is a transmembrane protein associated with metastatic characteristics of certain cancer types. However, it remains poorly characterized and its direct function in cancer remains unclear. The study presented here aims to further examine whether PTGFRN expression affects a cancer cell's phenotype, as well as metastatic-like characteristics. We used stable shRNA and cDNA transfections to respectively knockdown and overexpress PTGFRN in three different cancer cell lines, two of which are representative of rare and aggressive cancers (Mesothelioma and Pediatric Medulloblastoma). We then examined the characteristics of the resulting clones and showed a decrease in proliferation, migration, colony formation, and spheroid growth capabilities in cells where PTGFRN expression had been inhibited, while cells overexpressing PTGFRN showed the opposite. In addition, we showed that PTGFRN displayed direct binding to two protein partners, Integrin β1 and E. Cadherin, the latter of which is a novel direct binding partner to PTGFRN. Furthermore, silencing PTGFRN expression impacted the cellular process of autophagy, thereby providing another avenue by which PTGFRN potentially contributes to a cancer cell phenotype. Our findings demonstrate the potential role of PTGFRN in cancer metastasis and suggest PTGFRN as a future target for drug development in the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Jorge Marquez
- Department of Pharmaceutical Sciences, Baltimore School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Target Discovery Division, A&G Pharmaceutical Inc., Columbia, Maryland, USA
| | - Jianping Dong
- Target Discovery Division, A&G Pharmaceutical Inc., Columbia, Maryland, USA
| | - Jun Hayashi
- Precision Antibody Service, Columbia, Maryland, USA
| | - Ginette Serrero
- Target Discovery Division, A&G Pharmaceutical Inc., Columbia, Maryland, USA
- Precision Antibody Service, Columbia, Maryland, USA
| |
Collapse
|
12
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00969-z. [PMID: 39023664 DOI: 10.1007/s13402-024-00969-z] [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: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
13
|
Mrozowska M, Górnicki T, Olbromski M, Partyńska AI, Dzięgiel P, Rusak A. New insights into the role of tetraspanin 6, 7, and 8 in physiology and pathology. Cancer Med 2024; 13:e7390. [PMID: 39031113 PMCID: PMC11258570 DOI: 10.1002/cam4.7390] [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: 04/04/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND The tetraspanin (TSPAN) family comprises 33 membrane receptors involved in various physiological processes in humans. Tetrasapanins are surface proteins expressed in cells of various organisms. They are localised to the cell membrane by four transmembrane domains (TM4SF). These domains bind several cell surface receptors and signalling proteins to tetraspanin-enriched lipid microdomains (TERM or TEM). Tetraspanins play a critical role in anchoring many proteins. They also act as a scaffold for cell signalling proteins. AIM To summarise how tetraspanins 6, 7 and 8 contribute to the carcinogenesis process in different types of cancer. METHODS To provide a comprehensive review of the role of tetraspanins 6, 7 and 8 in cancer biology, we conducted a thorough search in PubMed, Embase and performed manual search of reference list to collect and extract data. DISCUSSION The assembly of tetraspanins covers an area of approximately 100-400 nm. Tetraspanins are involved in various biological processes such as membrane fusion, aggregation, proliferation, adhesion, cell migration and differentiation. They can also regulate integrins, cell surface receptors and signalling molecules. Tetraspanins form direct bonds with proteins and other members of the tetraspanin family, forming a hierarchical network of interactions and are thought to be involved in cell and membrane compartmentalisation. Tetraspanins have been implicated in cancer progression and have been shown to have multiple binding partners and to promote cancer progression and metastasis. Clinical studies have documented a correlation between the level of tetraspanin expression and the prediction of cancer progression, including breast and lung cancer. CONCLUSIONS Tetraspanins are understudied in almost all cell types and their functions are not clearly defined. Fortunately, it has been possible to identify the basic mechanisms underlying the biological role of these proteins. Therefore, the purpose of this review is to describe the roles of tetraspanins 6, 7 and 8.
Collapse
Affiliation(s)
- Monika Mrozowska
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
| | - Tomasz Górnicki
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
| | - Mateusz Olbromski
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
| | - Aleksandra Izabela Partyńska
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
- Department of Human Biology, Faculty of PhysiotherapyWroclaw University of Health and Sport SciencesWroclawPoland
| | - Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and EmbryologyWroclaw Medical UniversityWroclawPoland
| |
Collapse
|
14
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
15
|
Suwatthanarak T, Tanjak P, Chaiboonchoe A, Acharayothin O, Thanormjit K, Chanthercrob J, Suwatthanarak T, Niyomchan A, Tanaka M, Okochi M, Pongpaibul A, Chalermwai WV, Trakarnsanga A, Methasate A, Pithukpakorn M, Chinswangwatanakul V. Overexpression of TSPAN8 in consensus molecular subtype 3 colorectal cancer. Exp Mol Pathol 2024; 137:104911. [PMID: 38861838 DOI: 10.1016/j.yexmp.2024.104911] [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: 04/24/2023] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Recently, consensus molecular subtypes (CMSs) have been proposed as a robust transcriptome-based classification system for colorectal cancer (CRC). Tetraspanins (TSPANs) are transmembrane proteins. They have been associated with the development of numerous malignancies, including CRC, through their role as "master organizers" for multi-molecular membrane complexes. No previous study has investigated the correlation between TSPANs and CMS classification. Herein, we investigated the expression of TSPANs in patient-derived primary CRC tissues and their CMS classifications. METHODS RNA samples were derived from primary CRC tissues (n = 100 patients diagnosed with colorectal adenocarcinoma) and subjected to RNA sequencing for transcriptome-based CMS classification and TSPAN-relevant analyses. Immunohistochemistry (IHC) and immunofluorescence (IF) stains were conducted to observe the protein expression level. To evaluate the relative biological pathways, gene-set enrichment analysis was performed. RESULTS Of the highly expressed TSPAN genes in CRC tissues (TSPAN8, TSPAN29, and TSPAN30), TSPAN8 was notably overexpressed in CMS3-classified primary tissues. The overexpression of TSPAN8 protein in CMS3 CRC was also observed by IHC and IF staining. As a result of gene-set enrichment analysis, TSPAN8 may potentially play a role in organizing signaling complexes for kinase-based metabolic deregulation in CMS3 CRC. CONCLUSIONS The present study reports the overexpression of TSPAN8 in CMS3 CRC. This study proposes TSPAN8 as a subtype-specific biomarker for CMS3 CRC. This finding provides a foundation for future CMS-based studies of CRC, a complex disease and the second leading cause of cancer mortality worldwide.
Collapse
Affiliation(s)
- Thanawat Suwatthanarak
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pariyada Tanjak
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Systems Pharmacy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Onchira Acharayothin
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kullanist Thanormjit
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jantappapa Chanthercrob
- Siriraj Center of Systems Pharmacy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tharathorn Suwatthanarak
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Apichaya Niyomchan
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Kanagawa, Japan
| | - Mina Okochi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Tokyo, Japan
| | - Ananya Pongpaibul
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wipapat Vicki Chalermwai
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Atthaphorn Trakarnsanga
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Methasate
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Siriraj Center of Research Excellence in Precision Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vitoon Chinswangwatanakul
- Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
16
|
Atri Roozbahani G, Kokal-Ribaudo M, Heidari Horestani M, Pungsrinont T, Baniahmad A. The protein composition of exosomes released by prostate cancer cells is distinctly regulated by androgen receptor-antagonists and -agonist to stimulate growth of target cells. Cell Commun Signal 2024; 22:219. [PMID: 38589887 PMCID: PMC11000412 DOI: 10.1186/s12964-024-01584-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: 11/13/2023] [Accepted: 03/23/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a prevalent malignancy in men worldwide, ranking as the second leading cause of cancer-related death in Western countries. Various PCa hormone therapies, such as androgen receptor (AR)-antagonists or supraphysiological androgen level (SAL) reduce cancer cell proliferation. However, treated cells may influence the growth of neighboring cells through secreted exosomes in the tumor microenvironment (TME). Here, the change of protein content of exosomes secreted from PCa cells through treatment with different AR-antagonists or SAL has been analyzed. METHODS Isolation of exosomes via ultracentrifugation of treated human PCa LNCaP cells with AR-agonist and various AR-antagonists; analysis of cellular senescence by detection of senescence associated beta galactosidase activity (SA β-Gal); Western blotting and immunofluorescence staining; Mass spectrometry (MS-spec) of exosomes and bioinformatic analyses to identify ligand-specific exosomal proteins. Growth assays to analyze influence of exosomes on non-treated cells. RESULTS MS-spec analysis identified ligand-specific proteins in exosomes. One thousand seventy proteins were up- and 52 proteins downregulated by SAL whereas enzalutamide upregulated 151 proteins and downregulated 42 exosomal proteins. The bioinformatic prediction indicates an up-regulation of pro-proliferative pathways. AR ligands augment hub factors in exosomes that include AKT1, CALM1, PAK2 and CTNND1. Accordingly, functional assays confirmed that the isolated exosomes from AR-ligand treated cells promote growth of untreated PCa cells. CONCLUSION The data suggest that the cargo of exosomes is controlled by AR-agonist and -antagonists and distinct among the AR-antagonists. Further, exosomes promote growth that might influence the TME. This finding sheds light into the complex interplay between AR signaling and exosome-mediated communication between PCa cells.
Collapse
Affiliation(s)
- Golnaz Atri Roozbahani
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740, Jena, Germany
| | - Miriam Kokal-Ribaudo
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740, Jena, Germany
| | | | - Thanakorn Pungsrinont
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740, Jena, Germany
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07740, Jena, Germany.
| |
Collapse
|
17
|
Inyang I, White HE, Timme K, Keating AF. Biological sex differences in hepatic response to in utero dimethylbenz(a)anthracene exposure. Reprod Toxicol 2024; 124:108553. [PMID: 38307155 DOI: 10.1016/j.reprotox.2024.108553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
Fetal hepatic dimethylbenz(a)anthracene (DMBA) biotransformation is not defined, thus, this study investigated whether the fetal liver metabolizes DMBA and differs with biological sex. KK.Cg-a/a (lean; n = 20) or KK.Cg-Ay/J (obese; n = 20) pregnant mice were exposed to corn oil (CT) or DMBA (1 mg/kg bw/day) by intraperitoneal injection (n = 10/treatment) from gestation day 7-14. Postnatal day 2 male or female offspring livers were collected. Total RNA (n = 6) and protein (n = 6) were analyzed via a PCR-based array or LC-MS/MS, respectively. The level of Mgst3 was lower (P < 0.05) in livers of female compared to male offspring. Furthermore, in utero DMBA exposure increased (P < 0.1) Cyp2c29 and Gpx3 levels (P < 0.05) in female offspring. In male offspring, the abundance of Ahr, Comt (P < 0.1), Alox5, and Asna1 (P < 0.05) decreased due to DMBA exposure. Female and male offspring had 34 and 21 hepatic proteins altered (P < 0.05) by in utero DMBA exposure, respectively. Opposing patterns for hepatic CD81 and KRT78 occurred, being decreased in females but increased in males, while YWHAG was decreased by DMBA exposure in both. Functional KEGG pathway analysis identified enrichment of 26 and 13 hepatic metabolic proteins in male and female offspring, respectively, due to in utero DMBA exposure. In silico transcription factor analysis of differentially expressed proteins predicted involvement of female NRF1 but male AHR. Thus, hepatic biological sex differences and capacity to respond to toxicants in utero are supported.
Collapse
Affiliation(s)
| | - Hunter E White
- Department of Animal Science, Iowa State University, USA
| | - Kelsey Timme
- Department of Animal Science, Iowa State University, USA
| | | |
Collapse
|
18
|
Zhang C, Tang S, Wang M, Li L, Li J, Wang D, Mi X, Zhang Y, Tan X, Yue S. "Triple-Punch" Strategy Exosome-Mimetic Nanovesicles for Triple Negative Breast Cancer Therapy. ACS NANO 2024. [PMID: 38335265 DOI: 10.1021/acsnano.3c10568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most malignant breast cancer, with high rates of relapse and metastasis. Because of the nonspecific targeting of chemotherapy and insurmountable aggressiveness, TNBC therapy lacks an effective strategy. Exosomes have been reported as an efficient drug delivery system (DDS). CD82 is a tumor metastasis inhibitory molecule that is enriched in exosomes. Aptamer AS1411 specifically targets TNBC cells due to its high expression of nucleolin. We generated a "triple-punch" cell membrane-derived exosome-mimetic nanovesicle system that integrated with CD82 overexpression, AS1411 conjugation, and doxorubicin (DOX) delivery. CD82 enrichment effectively inhibits the migration of TNBC cells. AS1411 conjugation specifically targets TNBC cells. DOX loading effectively inhibits proliferation and induces apoptosis of TNBC cells. Our results demonstrate a system of exosome-mimetic nanovesicles with "triple-punch" that may facilitate TNBC therapeutics.
Collapse
Affiliation(s)
- Chenhong Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shuangshuang Tang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Luhan Li
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Li
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Dekun Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xue Mi
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
19
|
Runel G, Lopez-Ramirez N, Barbollat-Boutrand L, Cario M, Durand S, Grimont M, Schartl M, Dalle S, Caramel J, Chlasta J, Masse I. Cancer Cell Biomechanical Properties Accompany Tspan8-Dependent Cutaneous Melanoma Invasion. Cancers (Basel) 2024; 16:694. [PMID: 38398085 PMCID: PMC10887418 DOI: 10.3390/cancers16040694] [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: 12/17/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The intrinsic biomechanical properties of cancer cells remain poorly understood. To decipher whether cell stiffness modulation could increase melanoma cells' invasive capacity, we performed both in vitro and in vivo experiments exploring cell stiffness by atomic force microscopy (AFM). We correlated stiffness properties with cell morphology adaptation and the molecular mechanisms underlying epithelial-to-mesenchymal (EMT)-like phenotype switching. We found that melanoma cell stiffness reduction was systematically associated with the acquisition of invasive properties in cutaneous melanoma cell lines, human skin reconstructs, and Medaka fish developing spontaneous MAP-kinase-induced melanomas. We observed a systematic correlation of stiffness modulation with cell morphological changes towards mesenchymal characteristic gains. We accordingly found that inducing melanoma EMT switching by overexpressing the ZEB1 transcription factor, a major regulator of melanoma cell plasticity, was sufficient to decrease cell stiffness and transcriptionally induce tetraspanin-8-mediated dermal invasion. Moreover, ZEB1 expression correlated with Tspan8 expression in patient melanoma lesions. Our data suggest that intrinsic cell stiffness could be a highly relevant marker for human cutaneous melanoma development.
Collapse
Affiliation(s)
- Gaël Runel
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
- BioMeca, 60F, Bioserra 2, Av. Rockefeller, 69008 Lyon, France
| | - Noémie Lopez-Ramirez
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Laetitia Barbollat-Boutrand
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Muriel Cario
- National Reference Center for Rare Skin Disease, Department of Dermatology, University Hospital, INSERM 1035, 33000 Bordeaux, France
- AquiDerm, University Bordeaux, 33076 Bordeaux, France
| | - Simon Durand
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Maxime Grimont
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Manfred Schartl
- Developmental Biochemistry, University of Würzburg, 97074 Würzburg, Germany
- Xiphophorus Genetic Stock Center, Texas State University, San Marcos, TX 78666, USA
| | - Stéphane Dalle
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
- Dermatology Department, Hôpital Universitaire Lyon Sud, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Julie Caramel
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| | - Julien Chlasta
- BioMeca, 60F, Bioserra 2, Av. Rockefeller, 69008 Lyon, France
| | - Ingrid Masse
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, University Lyon 1, 69000 Lyon, France; (G.R.); (N.L.-R.)
| |
Collapse
|
20
|
Liu Y, Bai Y, Chen S, Pu F, Li Y, Chi H, Zheng Z, Xu P, Zhou T. Molecular characterization, expression pattern and immunologic function of CD82a in large yellow croaker ( Larimichthys crocea). Front Immunol 2024; 15:1301877. [PMID: 38370405 PMCID: PMC10869527 DOI: 10.3389/fimmu.2024.1301877] [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] [Received: 09/25/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Visceral white spot disease (VWND) caused by Pseudomonas plecoglossicida poses a major threat to the sustainable development of large yellow croaker (Larimichthys crocea) aquaculture. Genome-wide association analysis (GWAS) and RNA-seq research indicated that LcCD82a play an important role in resistance to visceral white spot disease in L. crocea, but the molecular mechanism of LcCD82a response to P. plecoglossicida infection is still unclear. In this study, we cloned and validated the Open Reading Frame (ORF) sequence of LcCD82a and explored the expression profile of LcCD82a in various tissues of L.crocea. In addition, two different transcript variants (LcCD82a-L and LcCD82a-S) of LcCD82a were identified that exhibit alternative splicing patterns after P. plecoglossicida infection, which may be closely related to the immune regulation during pathogenetic process of VWND. In order to explore the function of LcCD82a, we purified the recombinant protein of LcCD82a-L and LcCD82a-S. The bacterial agglutination and apoptosis function analysis showed that LcCD82a may involve in extracellular bacterial recognition, agglutination, and at the same time participate in the process of antigen presentation and induction of cell apoptosis. Collectively, our studies demonstrate that LcCD82a plays a crucial role in regulating apoptosis and antimicrobial immunity.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Yulin Bai
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Sijing Chen
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fei Pu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Yaxian Li
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Hongshu Chi
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Zaiyu Zheng
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Peng Xu
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Tao Zhou
- State Key Laboratory of Mariculture Breeding, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Genetics and Breeding of Marine Organisms, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
21
|
Leung KT, Cai J, Liu Y, Chan KYY, Shao J, Yang H, Hu Q, Xue Y, Wu X, Guo X, Zhai X, Wang N, Li X, Tian X, Li Z, Xue N, Guo Y, Wang L, Zou Y, Xiao P, He Y, Jin R, Tang J, Yang JJ, Shen S, Pui CH, Li CK. Prognostic implications of CD9 in childhood acute lymphoblastic leukemia: insights from a nationwide multicenter study in China. Leukemia 2024; 38:250-257. [PMID: 38001171 PMCID: PMC10844073 DOI: 10.1038/s41375-023-02089-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023]
Abstract
The outcomes of children with acute lymphoblastic leukemia (ALL) have been incrementally improved with risk-directed chemotherapy but therapy responses remain heterogeneous. Parameters with added prognostic values are warranted to refine the current risk stratification system and inform appropriate therapies. CD9, implicated by our prior single-center study, holds promise as one such parameter. To determine its precise prognostic significance, we analyzed a nationwide, multicenter, uniformly treated cohort of childhood ALL cases, where CD9 status was defined by flow cytometry on diagnostic samples of 3781 subjects. CD9 was expressed in 88.5% of B-ALL and 27.9% of T-ALL cases. It conferred a lower 5-year EFS and a higher CIR in B-ALL but not in T-ALL patients. The prognostic impact of CD9 was most pronounced in the intermediate/high-risk arms and those with minimal residual diseases, particularly at day 19 of remission induction. The adverse impact of CD9 was confined to specific cytogenetics, notably BCR::ABL1+ rather than KMT2A-rearranged leukemia. Multivariate analyses confirmed CD9 as an independent predictor of both events and relapse. The measurement of CD9 offers insights into patients necessitating intervention, warranting its seamless integration into the diagnostic marker panel to inform risk level and timely introduction of therapeutic intervention for childhood ALL.
Collapse
Affiliation(s)
- Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jiaoyang Cai
- Department of Hematology/Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, National Health Committee Key Laboratory of Pediatric Hematology & Oncology, Shanghai, China
| | - Yu Liu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, National Health Committee Key Laboratory of Pediatric Hematology & Oncology, Shanghai, China
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jingbo Shao
- Department of Hematology/Oncology, Shanghai Children's Hospital, Shanghai, China
| | - Hui Yang
- Department of Pediatrics, Xiangya Hospital Central South University, Changsha, China
| | - Qun Hu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Xue
- Department of Hematology/Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xia Guo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, Chengdu, China
| | - Xiaowen Zhai
- Department of Hematology/Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Ningling Wang
- Department of Pediatrics, Anhui Medical University Second Affiliated Hospital, Anhui, China
| | - Xue Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Tian
- Department of Hematology/Oncology, KunMing Children's Hospital, Kunming, China
| | - Zheng Li
- Department of Hematology/Oncology, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Ning Xue
- Department of Hematology/Oncology, Xi 'an Northwest Women's and Children's Hospital, Xi 'an, China
| | - Yuxia Guo
- Department of Hematology/Oncology, Chongqing Medical University Affiliated Children's Hospital, Chongqing, China
| | - Lingzhen Wang
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yao Zou
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Peifang Xiao
- Department of Hematology/Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Yingyi He
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyan Tang
- Department of Hematology/Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, National Health Committee Key Laboratory of Pediatric Hematology & Oncology, Shanghai, China
| | - Jun J Yang
- Departments of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shuhong Shen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, National Health Committee Key Laboratory of Pediatric Hematology & Oncology, Shanghai, China.
| | - Ching-Hon Pui
- Departments of Oncology, Pathology, and Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Chi Kong Li
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
22
|
Yang J, Zhang Z, Lam JSW, Fan H, Fu NY. Molecular Regulation and Oncogenic Functions of TSPAN8. Cells 2024; 13:193. [PMID: 38275818 PMCID: PMC10814125 DOI: 10.3390/cells13020193] [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: 01/03/2024] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Tetraspanins, a superfamily of small integral membrane proteins, are characterized by four transmembrane domains and conserved protein motifs that are configured into a unique molecular topology and structure in the plasma membrane. They act as key organizers of the plasma membrane, orchestrating the formation of specialized microdomains called "tetraspanin-enriched microdomains (TEMs)" or "tetraspanin nanodomains" that are essential for mediating diverse biological processes. TSPAN8 is one of the earliest identified tetraspanin members. It is known to interact with a wide range of molecular partners in different cellular contexts and regulate diverse molecular and cellular events at the plasma membrane, including cell adhesion, migration, invasion, signal transduction, and exosome biogenesis. The functions of cell-surface TSPAN8 are governed by ER targeting, modifications at the Golgi apparatus and dynamic trafficking. Intriguingly, limited evidence shows that TSPAN8 can translocate to the nucleus to act as a transcriptional regulator. The transcription of TSPAN8 is tightly regulated and restricted to defined cell lineages, where it can serve as a molecular marker of stem/progenitor cells in certain normal tissues as well as tumors. Importantly, the oncogenic roles of TSPAN8 in tumor development and cancer metastasis have gained prominence in recent decades. Here, we comprehensively review the current knowledge on the molecular characteristics and regulatory mechanisms defining TSPAN8 functions, and discuss the potential and significance of TSPAN8 as a biomarker and therapeutic target across various epithelial cancers.
Collapse
Affiliation(s)
- Jicheng Yang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ziyan Zhang
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Joanne Shi Woon Lam
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore
| | - Hao Fan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore
| | - Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
23
|
Zhang XM, Huang J, Ni XY, Zhu HR, Huang ZX, Ding S, Yang XY, Tan YD, Chen JF, Cai JH. Current progression in application of extracellular vesicles in central nervous system diseases. Eur J Med Res 2024; 29:15. [PMID: 38173021 PMCID: PMC10763486 DOI: 10.1186/s40001-023-01606-5] [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: 04/11/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Early diagnosis and pharmacological treatment of central nervous system (CNS) diseases has been a long-standing challenge for clinical research due to the presence of the blood-brain barrier. Specific proteins and RNAs in brain-derived extracellular vesicles (EVs) usually reflect the corresponding state of brain disease, and therefore, EVs can be used as diagnostic biomarkers for CNS diseases. In addition, EVs can be engineered and fused to target cells for delivery of cargo, demonstrating the great potential of EVs as a nanocarrier platform. We review the progress of EVs as markers and drug carriers in the diagnosis and treatment of neurological diseases. The main areas include visual imaging, biomarker diagnosis and drug loading therapy for different types of CNS diseases. It is hoped that increased knowledge of EVs will facilitate their clinical translation in CNS diseases.
Collapse
Affiliation(s)
- Xiang-Min Zhang
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Jie Huang
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Xiao-Ying Ni
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Hui-Ru Zhu
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Zhong-Xin Huang
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Shuang Ding
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Xin-Yi Yang
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, No. 136, Zhongshan Second Road, Chongqing, 400014, China
| | - Yan-Di Tan
- Department of Ultrasound the Third Affiliated Hospital of Chongqing Medical University, No. 1, Shuanghu Branch Road, Huixing Street, Chongqing, 401120, China
| | - Jian-Fu Chen
- Department of Ultrasound, The Second People's Hospital of Yunnan Province, No. 176, Qingnian Road, Kunming, 650021, China
| | - Jin-Hua Cai
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, No. 136, Zhongshan Second Road, Chongqing, 400014, China.
| |
Collapse
|
24
|
Nazri HM, Greaves E, Quenby S, Dragovic R, Tapmeier TT, Becker CM. The role of small extracellular vesicle-miRNAs in endometriosis. Hum Reprod 2023; 38:2296-2311. [PMID: 37877421 PMCID: PMC10694411 DOI: 10.1093/humrep/dead216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Endometriosis is defined by the presence of extrauterine endometrial-like tissue, which can cause pain and infertility in 10% of reproductive-age women. To date, the pathogenesis is poorly understood resulting in significant diagnostic delays and poor therapeutic outcomes in many women. Small extracellular vesicles (sEVs) (<200 nm) are cell-derived vesicles containing molecules that can influence gene expression and behaviour in target cells. One such cargo are microRNAs (miRNAs), which are short, non-coding RNAs mostly 19-25 nucleotides in length that regulate post-transcriptional gene expression. This mini-review focuses on the role of sEV-miRNAs, which are conceivably better biomarkers for endometriosis than free miRNAs, which reflect the true pathophysiological state in the body, as sEV-encapsulated miRNAs are protected from degradation compared to free miRNA and provide direct cell-to-cell communication via sEV surface proteins. sEV-miRNAs have been implicated in the immunomodulation of macrophages, the proliferation, migration and invasion of endometrial cells, and angiogenesis, all hallmarks of endometriosis. The diagnostic potential of sEV-miRNA was investigated in one study that reported the sensitivity and specificity of two sEV-miRNAs (hsa-miR-22-3p and hsa-miR-320a-3p) in distinguishing endometriosis from non-endometriosis cases. Only three studies have explored the therapeutic potential of sEV-miRNAs in vivo in mice-two looked into the role of sEV-hsa-miR-214-3p in decreasing fibrosis, and one investigated sEV-hsa-miR-30c-5p in suppressing the invasive and migratory potential of endometriotic lesions. While early results are encouraging, studies need to further address the potential influence of factors such as the menstrual cycle as well as the location and extent of endometriotic lesions on miRNA expression in sEVs. Given these findings, and extrapolating from other conditions such as cancer, diabetes, and pre-eclampsia, sEV-miRNAs could present an attractive and urgently needed future diagnostic and therapeutic target for millions of women suffering from endometriosis. However, research in this area is hampered by lack of adherence to the International Society for Extracellular Vesicles 2018 guideline in separating and characterising sEVs, as well as the World Endometriosis Research Foundation Endometriosis Phenome and Biobanking Harmonisation Project protocols.
Collapse
Affiliation(s)
- Hannah M Nazri
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Siobhan Quenby
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Rebecca Dragovic
- Nuffield Department of Women’s & Reproductive Health, Endometriosis CaRe Centre, University of Oxford, Oxford, UK
| | - Thomas T Tapmeier
- Nuffield Department of Women’s & Reproductive Health, Endometriosis CaRe Centre, University of Oxford, Oxford, UK
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Christian M Becker
- Nuffield Department of Women’s & Reproductive Health, Endometriosis CaRe Centre, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Szatmári T, Balázs K, Csordás IB, Sáfrány G, Lumniczky K. Effect of radiotherapy on the DNA cargo and cellular uptake mechanisms of extracellular vesicles. Strahlenther Onkol 2023; 199:1191-1213. [PMID: 37347291 DOI: 10.1007/s00066-023-02098-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/01/2023] [Indexed: 06/23/2023]
Abstract
In the past decades, plenty of evidence has gathered pointing to the role of extracellular vesicles (EVs) secreted by irradiated cells in the development of radiation-induced non-targeted effects. EVs are complex natural structures composed of a phospholipid bilayer which are secreted by virtually all cells and carry bioactive molecules. They can travel certain distances in the body before being taken up by recipient cells. In this review we discuss the role and fate of EVs in tumor cells and highlight the importance of DNA specimens in EVs cargo in the context of radiotherapy. The effect of EVs depends on their cargo, which reflects physiological and pathological conditions of donor cell types, but also depends on the mode of EV uptake and mechanisms involved in the route of EV internalization. While the secretion and cargo of EVs from irradiated cells has been extensively studied in recent years, their uptake is much less understood. In this review, we will focus on recent knowledge regarding the EV uptake of cancer cells and the effect of radiation in this process.
Collapse
Affiliation(s)
- Tünde Szatmári
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary.
| | - Katalin Balázs
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Ilona Barbara Csordás
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Géza Sáfrány
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| | - Katalin Lumniczky
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, 1097, Budapest, Hungary
| |
Collapse
|
26
|
da Silva TB, Rendra E, David CAW, Bieback K, Cross MJ, Wilm B, Liptrott NJ, Murray P. Umbilical cord mesenchymal stromal cell-derived extracellular vesicles lack the potency to immunomodulate human monocyte-derived macrophages in vitro. Biomed Pharmacother 2023; 167:115624. [PMID: 37783151 DOI: 10.1016/j.biopha.2023.115624] [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: 07/01/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been reported to display efficacy in a variety of preclinical models, but without long-term engraftment, suggesting a role for secreted factors, such as MSC-derived extracellular vesicles (EVs). MSCs are known to elicit immunomodulatory effects, an important aspect of which is their ability to affect macrophage phenotype. However, it is not clear if these effects are mediated by MSC-derived EVs, or other factors secreted by the MSCs. Here, we use flow cytometry to assess the effects of human umbilical cord (hUC) MSC-derived EVs on the expression of pro-inflammatory (CD80) and anti-inflammatory (CD163) surface markers in human monocyte-derived macrophages (hMDMs). hUC-MSC-derived EVs did not change the surface marker expression of the hMDMs. In contrast, when hMDMs were co-incubated with hUC-MSCs in indirect co-cultures, changes were observed in the expression of CD14, CD80 and CD163, particularly in M1 macrophages, suggesting that soluble factors are necessary to elicit a shift in phenotype. However, even though EVs did not alter the surface marker expression of macrophages, they promoted angiogenesis and phagocytic capacity increased proportionally to increases in EV concentration. Taken together, these results suggest that hUC-MSC-derived EVs are not sufficient to alter macrophage phenotype and that additional MSC-derived factors are needed.
Collapse
Affiliation(s)
- Tamiris Borges da Silva
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK
| | - Erika Rendra
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Christopher A W David
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Michael J Cross
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK
| | - Neill J Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, L69 3GE Liverpool, UK.
| |
Collapse
|
27
|
Das K, Paul S, Ghosh A, Gupta S, Mukherjee T, Shankar P, Sharma A, Keshava S, Chauhan SC, Kashyap VK, Parashar D. Extracellular Vesicles in Triple-Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers (Basel) 2023; 15:4879. [PMID: 37835573 PMCID: PMC10571545 DOI: 10.3390/cancers15194879] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype accounting for ~10-20% of all human BC and is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) amplification. Owing to its unique molecular profile and limited targeted therapies, TNBC treatment poses significant challenges. Unlike other BC subtypes, TNBC lacks specific molecular targets, rendering endocrine therapies and HER2-targeted treatments ineffective. The chemotherapeutic regimen is the predominant systemic treatment modality for TNBC in current clinical practice. However, the efficacy of chemotherapy in TNBC is variable, with response rates varying between a wide range of patients, and the emerging resistance further adds to the difficulties. Furthermore, TNBC exhibits a higher mutational burden and is acknowledged as the most immunogenic of all BC subtypes. Consequently, the application of immune checkpoint inhibition has been investigated in TNBC, yielding promising outcomes. Recent evidence identified extracellular vesicles (EVs) as an important contributor in the context of TNBC immunotherapy. In view of the extraordinary ability of EVs to transfer bioactive molecules, such as proteins, lipids, DNA, mRNAs, and small miRNAs, between the cells, EVs are considered a promising diagnostic biomarker and novel drug delivery system among the prospects for immunotherapy. The present review provides an in-depth understanding of how EVs influence TNBC progression, its immune regulation, and their contribution as a predictive biomarker for TNBC. The final part of the review focuses on the recent key advances in immunotherapeutic strategies for better understanding the complex interplay between EVs and the immune system in TNBC and further developing EV-based targeted immunotherapies.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, Galveston, TX 77555, USA or
| | - Anshul Sharma
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Vivek Kumar Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
28
|
El-Maradny YA, Rubio-Casillas A, Mohamed KI, Uversky VN, Redwan EM. Intrinsic factors behind long-COVID: II. SARS-CoV-2, extracellular vesicles, and neurological disorders. J Cell Biochem 2023; 124:1466-1485. [PMID: 37801299 DOI: 10.1002/jcb.30486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
With the decline in the number of new Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infections, the World Health Organization announced the end of the SARS-CoV-2 pandemic. However, the repercussions of this viral pandemic may remain with us for a longer period of time, as it has remodeled the lives of humankind in many ways, including social and economic. Of course, its most important repercussions remain on the human health level. Long-coronavirus disease (COVID) or post-COVID is a state for which we do not have a concrete definition, a specific international classification of diseases Code, clear diagnostic tools, or well-known effective cures as of yet. In this second article from the Intrinsic Factors behind long-COVID Series, we try to link long-COVID symptoms with their causes, starting from the nervous system. Extracellular vesicles (ECVs) play very complex and ramified roles in the bodies of both healthy and not-healthy individuals. ECVs may facilitate the entry of many bioactive molecules and pathogens into the tissues and cells of the nervous system across the blood-brain barrier. Based on the size, quantity, and quality of their cargo, ECVs are directly proportional to the pathological condition and its severity through intertwined mechanisms that evoke inflammatory immune responses typically accompanied by pathological symptoms over variable time periods according to the type of these symptoms.
Collapse
Affiliation(s)
- Yousra A El-Maradny
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Egypt
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein, Egypt
| | - Alberto Rubio-Casillas
- Biology Laboratory, Autlán Regional Preparatory School, University of Guadalajara, Autlán, Jalisco, Mexico
| | - Kareem I Mohamed
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Zhu Y, Saint-Pol J, Nguyen V, Rubinstein E, Boucheix C, Greco C. The Tetraspanin Tspan8 Associates with Endothelin Converting Enzyme ECE1 and Regulates Its Activity. Cancers (Basel) 2023; 15:4751. [PMID: 37835445 PMCID: PMC10571763 DOI: 10.3390/cancers15194751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Tspan8 is a member of the tetraspanins family of cell surface molecules. The ability of tetraspanins to organize membrane microdomains with other membrane molecules and interfere with their function suggests that they could act as surface integrators of external or internal signals. Among the first identified tetraspanins, Tspan8 promotes tumor progression and metastasis, presumably by stimulating angiogenesis and cell motility. In patients, its expression on digestive tract tumors seems to be associated with a bad prognosis. We showed previously that Tspan8 associates with E-cadherin and EGFR and modulates their effects on cell motility. Using Mass spectrometry and western blot, we found a new partner, the endothelin converting enzyme ECE1, and showed that Tspan8 amplifies its activity of conversion of the endothelin-1 precursor bigET1 to endothelin. This was observed by transduction of the colon carcinoma cell line Isreco1, which does not express Tspan8, and on ileum tissue fragments of tspan8ko mice versus wild type mice. Given these results, Tspan8 appears to be a modulator of the endothelin axis, which could possibly be targeted in case of over-activity of endothelins in biological processes of tissues expressing Tspan8.
Collapse
Affiliation(s)
- Yingying Zhu
- UMR-S 1004, SFR André Lwoff, Inserm, 94800 Villejuif, France; (Y.Z.); (J.S.-P.); (E.R.)
- Université Paris Saclay, 91190 Gif-sur-Yvette, France;
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Julien Saint-Pol
- UMR-S 1004, SFR André Lwoff, Inserm, 94800 Villejuif, France; (Y.Z.); (J.S.-P.); (E.R.)
- Université Artois, UR 2465, Blood-Brain Barrier Laboratory (LBHE), 62300 Lens, France
| | - Viet Nguyen
- Université Paris Saclay, 91190 Gif-sur-Yvette, France;
- Plateforme Spectrométrie de Masse, Laboratoire Biochimie-Hormonologie, Hôpital Robert-Debré, Assistance Publique Hôpitaux de Paris (APHP), 75610 Paris, France
| | - Eric Rubinstein
- UMR-S 1004, SFR André Lwoff, Inserm, 94800 Villejuif, France; (Y.Z.); (J.S.-P.); (E.R.)
- Centre d’Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, CIMI-Paris, 75013 Paris, France
| | - Claude Boucheix
- Université Paris Saclay, 91190 Gif-sur-Yvette, France;
- UMR-S 1197, SFR André Lwoff, Inserm, 94800 Villejuif, France
| | - Céline Greco
- Department of Pain and Palliative Care, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (APHP), 75610 Paris, France
- U1163, IMAGINE Institute, Université de Paris Cité, Inserm, 75014 Paris, France
| |
Collapse
|
30
|
Zhang W, Ling Y, Sun Y, Xiao F, Wang L. Extracellular Vesicles Derived from Mesenchymal Stem Cells Promote Wound Healing and Skin Regeneration by Modulating Multiple Cellular Changes: A Brief Review. Genes (Basel) 2023; 14:1516. [PMID: 37628568 PMCID: PMC10453884 DOI: 10.3390/genes14081516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are biologically active substances secreted by MSCs into the extracellular matrix that play an immunomodulatory role in skin damage repair. To investigate the mechanism of MSC-EVs in reducing inflammation, promoting angiogenesis, promoting the proliferation and migration of epithelial cells and fibroblasts, and extracellular matrix remodeling during wound healing, we focused on the effects of EVs on multiple cell types at various stages of skin injury. A literature review was conducted to explore related research on the influence of MSC-EVs on the types of cells involved in wound healing. MSC-EVs show a strong regulatory ability on immune cells involved in the regulation of inflammation, including macrophages, neutrophils, and T cells, and other cells involved in tissue proliferation and remodeling, such as fibroblasts, keratinocytes, and endothelial cells, during wound healing in in vitro and in vivo experiments, which substantially promoted the understanding of wound healing in the field of trauma medicine. MSC-EVs have potential applications in combating poor skin wound healing. Elucidating the mechanism of action of EVs in the wound-healing process would greatly advance the understanding of therapeutic wound healing.
Collapse
Affiliation(s)
- Weiyuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (W.Z.); (Y.L.); (Y.S.)
| | - Yang Ling
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (W.Z.); (Y.L.); (Y.S.)
| | - Yang Sun
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (W.Z.); (Y.L.); (Y.S.)
| | - Fengjun Xiao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lisheng Wang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao 266000, China; (W.Z.); (Y.L.); (Y.S.)
| |
Collapse
|
31
|
Parra-Aguilar TJ, Sarmiento-López LG, Santana O, Olivares JE, Pascual-Morales E, Jiménez-Jiménez S, Quero-Hostos A, Palacios-Martínez J, Chávez-Martínez AI, Cárdenas L. TETRASPANIN 8-1 from Phaseolus vulgaris plays a key role during mutualistic interactions. FRONTIERS IN PLANT SCIENCE 2023; 14:1152493. [PMID: 37465390 PMCID: PMC10352089 DOI: 10.3389/fpls.2023.1152493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/13/2023] [Indexed: 07/20/2023]
Abstract
Arbuscular mycorrhizal (AM) fungi and rhizobia form two of the most important plant-microbe associations for the assimilation of phosphorus (P) and nitrogen (N). Symbiont-derived signals are able to coordinate the infection process by triggering multiple responses in the plant root, such as calcium influxes and oscillations, increased reactive oxygen species (ROS), cytoskeletal rearrangements and altered gene expression. An examination was made of the role of tetraspanins, which are transmembrane proteins that self-organize into tetraspanin web regions, where they recruit specific proteins into platforms required for signal transduction, membrane fusion, cell trafficking, and ROS generation. In plant cells, tetraspanins are scaffolding proteins associated with root radial patterning, biotic and abiotic stress responses, cell fate determination, plasmodesmata and hormonal regulation. Some plant tetraspanins, such as Arabidopsis thaliana TETRASPANIN 8 and TETRASPANIN 9 (AtTET8 and AtTET9) are associated with exosomes during inter-kingdom communication. In this study, a homolog of AtTET8, PvTET8-1, in common bean (Phaseolus vulgaris L. var. Negro Jamapa) was examined in roots during interactions with Rhizobium tropici and Rhizophagus irregularis. The promoter of PvTET8-1 contained several cis-acting regulatory DNA elements potentially related to mutualistic interactions, and PvTET8-1 was transcriptionally activated during AM fungal and rhizobial associations. Silencing it decreased the size and number of nodules, nitrogen fixation, and mycorrhizal arbuscule formation, whereas overexpressing it increased the size and number of nodules, and mycorrhizal arbuscule formation but decreased nitrogen fixation. PvTET8-1 appears to be an important element in both of these mutualistic interactions, perhaps through its interaction with NADPH oxidase and the generation of ROS during the infection processes.
Collapse
Affiliation(s)
- Thelma J. Parra-Aguilar
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Luis G. Sarmiento-López
- Departamento de Biotecnología Agrícola, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional Unidad Sinaloa-Instituto Politécnico Nacional, Guasave, Sinaloa, Mexico
| | - Olivia Santana
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Juan Elías Olivares
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Edgar Pascual-Morales
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Saul Jiménez-Jiménez
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Andrea Quero-Hostos
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Janet Palacios-Martínez
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Ana I. Chávez-Martínez
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Luis Cárdenas
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
32
|
Das K, Mukherjee T, Shankar P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules 2023; 13:897. [PMID: 37371477 DOI: 10.3390/biom13060897] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
The tumor microenvironment (TME) plays an important role in the development and progression of hematological malignancies. In recent years, studies have focused on understanding how tumor cells communicate within the TME. In addition to several factors, such as growth factors, cytokines, extracellular matrix (ECM) molecules, etc., a growing body of evidence has indicated that extracellular vesicles (EVs) play a crucial role in the communication of tumor cells within the TME, thereby contributing to the pathogenesis of hematological malignancies. The present review focuses on how EVs derived from tumor cells interact with the cells in the TME, such as immune cells, stromal cells, endothelial cells, and ECM components, and vice versa, in the context of various hematological malignancies. EVs recovered from the body fluids of cancer patients often carry the bioactive molecules of the originating cells and hence can be considered new predictive biomarkers for specific types of cancer, thereby also acting as potential therapeutic targets. Here, we discuss how EVs influence hematological tumor progression via tumor-host crosstalk and their use as biomarkers for hematological malignancies, thereby benefiting the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Tanmoy Mukherjee
- Department of Pulmonary Immunology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Prem Shankar
- Department of Pulmonary Immunology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| |
Collapse
|
33
|
Yang J, Guo F, Chin HS, Chen GB, Ang CH, Lin Q, Hong W, Fu NY. Sequential genome-wide CRISPR-Cas9 screens identify genes regulating cell-surface expression of tetraspanins. Cell Rep 2023; 42:112065. [PMID: 36724073 DOI: 10.1016/j.celrep.2023.112065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/16/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Tetraspanins, a superfamily of membrane proteins, mediate diverse biological processes through tetraspanin-enriched microdomains in the plasma membrane. However, how their cell-surface presentation is controlled remains unclear. To identify the regulators of tetraspanin trafficking, we conduct sequential genome-wide loss-of-function CRISPR-Cas9 screens based on cell-surface expression of a tetraspanin member, TSPAN8. Several genes potentially involved in endoplasmic reticulum (ER) targeting, different biological processes in the Golgi apparatus, and protein trafficking are identified and functionally validated. Importantly, we find that biantennary N-glycans generated by MGAT1/2, but not more complex glycan structures, are important for cell-surface tetraspanin expression. Moreover, we unravel that SPPL3, a Golgi intramembrane-cleaving protease reported previously to act as a sheddase of multiple glycan-modifying enzymes, controls cell-surface tetraspanin expression through a mechanism associated with lacto-series glycolipid biosynthesis. Our study provides critical insights into the molecular regulation of cell-surface presentation of tetraspanins with implications for strategies to manipulate their functions, including cancer cell invasion.
Collapse
Affiliation(s)
- Jicheng Yang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Fusheng Guo
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Hui San Chin
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Gao Bin Chen
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Chow Hiang Ang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A(∗)STAR), Singapore 138673, Singapore
| | - Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Physiology, National University of Singapore, Singapore 117593, Singapore; Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
34
|
Sanz-Ros J, Mas-Bargues C, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Extracellular Vesicles as Therapeutic Resources in the Clinical Environment. Int J Mol Sci 2023; 24:2344. [PMID: 36768664 PMCID: PMC9917082 DOI: 10.3390/ijms24032344] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
The native role of extracellular vesicles (EVs) in mediating the transfer of biomolecules between cells has raised the possibility to use them as therapeutic vehicles. The development of therapies based on EVs is now expanding rapidly; here we will describe the current knowledge on different key points regarding the use of EVs in a clinical setting. These points are related to cell sources of EVs, isolation, storage, and delivery methods, as well as modifications to the releasing cells for improved production of EVs. Finally, we will depict the application of EVs therapies in clinical trials, considering the impact of the COVID-19 pandemic on the development of these therapies, pointing out that although it is a promising therapy for human diseases, we are still in the initial phase of its application to patients.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Department of Cardiology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari de Valencia, University of Valencia, 46010 Valencia, Spain
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
35
|
Chen T, Wang F, Wei J, Feng L. Extracellular vesicles derived from different sources play various roles in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 13:1064415. [PMID: 36686474 PMCID: PMC9845915 DOI: 10.3389/fendo.2022.1064415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023] Open
Abstract
Extracellular vesicles (EVs) are present in almost all biological fluids and secreted by almost all cell types. A growing number of studies have revealed the potential roles of EVs in the diagnosis and treatment of the diabetic retinopathy (DR). Changes in the quantity and content of EVs may serve as biomarkers of cause or consequence of pathological status of DR, such as inflammation, neovascularization and epithelial-mesenchymal transition. In addition, as natural, safe and efficient drug carrier, EVs have been reported to play important roles in intercellular communication by acting for essential cell-specific information to target cells. In this review, we summarize the roles of EVs, secreted by various types of cells and participated in various biological processes, in the pathogenesis, diagnosis, and treatment of DR.
Collapse
Affiliation(s)
| | | | | | - Le Feng
- Department of Ophthalmology, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Tan M, Ge Y, Wang X, Wang Y, Liu Y, He F, Teng H. Extracellular Vesicles (EVs) in Tumor Diagnosis and Therapy. Technol Cancer Res Treat 2023; 22:15330338231171463. [PMID: 37122245 PMCID: PMC10134167 DOI: 10.1177/15330338231171463] [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: 05/02/2023] Open
Abstract
In recent years, extracellular vesicles (EVs) have gained significant attention due to their tremendous potential for clinical applications. EVs play a crucial role in various aspects, including tumorigenesis, drug resistance, immune escape, and reconstruction of the tumor microenvironment. Despite the growing interest in EVs, many questions still need to be addressed before they can be practically applied in clinical settings. This paper aims to review EVs' isolation methods, structure research, the roles of EVs in tumorigenesis and their mechanisms in multiple types of tumors, their potential application in drug delivery, and the expectations for their future in clinical research.
Collapse
Affiliation(s)
- Mingdian Tan
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Yizhi Ge
- The Affiliated Cancer Hospital of Nanjing Medical University (Jiangsu Cancer Hospital) and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiaogang Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yan Wang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Liu
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Feng He
- Stanford University School of Medicine, Stanford, CA, USA
| | - Hongqi Teng
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
37
|
Cheng P, Wang X, Liu Q, Yang T, Qu H, Zhou H. Extracellular vesicles mediate biological information delivery: A double-edged sword in cardiac remodeling after myocardial infarction. Front Pharmacol 2023; 14:1067992. [PMID: 36909157 PMCID: PMC9992194 DOI: 10.3389/fphar.2023.1067992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Acute myocardial infarction (AMI) is a severe ischemic disease with high morbidity and mortality worldwide. Maladaptive cardiac remodeling is a series of abnormalities in cardiac structure and function that occurs following myocardial infarction (MI). The pathophysiology of this process can be separated into two distinct phases: the initial inflammatory response, and the subsequent longer-term scar revision that includes the regression of inflammation, neovascularization, and fibrotic scar formation. Extracellular vesicles are nano-sized lipid bilayer vesicles released into the extracellular environment by eukaryotic cells, containing bioinformatic transmitters which are essential mediators of intercellular communication. EVs of different cellular origins play an essential role in cardiac remodeling after myocardial infarction. In this review, we first introduce the pathophysiology of post-infarction cardiac remodeling, as well as the biogenesis, classification, delivery, and functions of EVs. Then, we explore the dual role of these small molecule transmitters delivered by EVs in post-infarction cardiac remodeling, including the double-edged sword of pro-and anti-inflammation, and pro-and anti-fibrosis, which is significant for post-infarction cardiac repair. Finally, we discuss the pharmacological and engineered targeting of EVs for promoting heart repair after MI, thus revealing the potential value of targeted modulation of EVs and its use as a drug delivery vehicle in the therapeutic process of post-infarction cardiac remodeling.
Collapse
Affiliation(s)
- Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Yang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huiyan Qu
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
38
|
Kim SD, Cho KS. Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease. LIFE (BASEL, SWITZERLAND) 2022; 12:life12121994. [PMID: 36556359 PMCID: PMC9786036 DOI: 10.3390/life12121994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
Mesenchymal stem cells (MSCs) have been reported as promising candidates for the treatment of various diseases, especially allergic diseases, as they have the capacity to differentiate into various cells. However, MSCs itself have several limitations such as creating a risk of aneuploidy, difficulty in handling them, immune rejection, and tumorigenicity, so interest in the extracellular vesicles (EVs) released from MSCs are increasing, and many studies have been reported. Previous studies have shown that extracellular vesicles (EVs) produced by MSCs are as effective as the MSCs themselves in suppression of allergic airway inflammation through the suppression of Th2 cytokine production and the induction of regulatory T cells (Treg) expansion. EVs are one of the substances secreted by paracrine induction from MSCs, and because it exerts its effect by delivering contents such as mRNA, microRNA, and proteins to the receptor cell, it can reduce the problems or risks related to stem cell therapy. This article reviews the immunomodulatory properties of MSCs-derived EVs and their therapeutic implications for allergic airway disease.
Collapse
Affiliation(s)
- Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan 602-739, Republic of Korea
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan 602-739, Republic of Korea
| |
Collapse
|
39
|
Co-expression of Matrix Metalloproteinase 9 (MMP9) and Motility-related Protein-1 (MRP-1/CD9) in Human Breast Cancer. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm-105009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Controversial evidence exists regarding the metastasis-suppressor or promoting activity of motility-related protein-1 (MRP-1/CD9) in breast cancer (BC). Objectives: we aimed at investigating the possible correlation of CD9 with a metastasis marker matrix metalloproteinase 9 (MMP9) in human BC tissues. Methods: A total of 19 BC and 5 normal breast tissues were analyzed. The mRNA expression of CD9 and MMP9 was measured by quantitative PCR. The correlation of genes’ expression with each other and clinicopathological features (i. e. tumor pathology, grade, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER-2), P53, and KI67) was tested by relevant statistical analysis methods. Results: There was no significant difference between patients with BC and the control group regarding the expression of MMP9 (P = 0.394) and CD9 (P = 0.887). A significant strong positive correlation was observed between CD9 and MMP-9 expressions (r = 0.761 and P = 0.0002). The MMP9 expression was significantly higher in ER or PR positive compared to ER or PR negative tumors (P = 0.046). Conclusions: Given the strong correlation with MMP9, it seems that CD9 might have a role in metastasis in human BC. However, much more studies are required to further support this hypothesis.
Collapse
|
40
|
Sanz-Ros J, Romero-García N, Mas-Bargues C, Monleón D, Gordevicius J, Brooke RT, Dromant M, Díaz A, Derevyanko A, Guío-Carrión A, Román-Domínguez A, Inglés M, Blasco MA, Horvath S, Viña J, Borrás C. Small extracellular vesicles from young adipose-derived stem cells prevent frailty, improve health span, and decrease epigenetic age in old mice. SCIENCE ADVANCES 2022; 8:eabq2226. [PMID: 36260670 PMCID: PMC9581480 DOI: 10.1126/sciadv.abq2226] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Aging is associated with an increased risk of frailty, disability, and mortality. Strategies to delay the degenerative changes associated with aging and frailty are particularly interesting. We treated old animals with small extracellular vesicles (sEVs) derived from adipose mesenchymal stem cells (ADSCs) of young animals, and we found an improvement in several parameters usually altered with aging, such as motor coordination, grip strength, fatigue resistance, fur regeneration, and renal function, as well as an important decrease in frailty. ADSC-sEVs induced proregenerative effects and a decrease in oxidative stress, inflammation, and senescence markers in muscle and kidney. Moreover, predicted epigenetic age was lower in tissues of old mice treated with ADSC-sEVs and their metabolome changed to a youth-like pattern. Last, we gained some insight into the microRNAs contained in sEVs that might be responsible for the observed effects. We propose that young sEV treatment can promote healthy aging.
Collapse
Affiliation(s)
- Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Daniel Monleón
- Department of Pathology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia, Spain
| | | | | | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Ana Díaz
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Aksinya Derevyanko
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, 28029 Madrid, Spain
| | - Ana Guío-Carrión
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, 28029 Madrid, Spain
| | - Aurora Román-Domínguez
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Marta Inglés
- Freshage Research Group, Department of Physiotherapy, Faculty of Physiotherapy, University of Valencia, CIBERFES, INCLIVA, Avenida Blasco Ibáñez, 15, Valencia Spain
| | - María A. Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, 28029 Madrid, Spain
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Jose Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Corresponding author.
| |
Collapse
|
41
|
Ghatak S, Hascall VC, Karamanos N, Markwald RR, Misra S. Chemotherapy induces feedback up-regulation of CD44v6 in colorectal cancer initiating cells through β-catenin/MDR1 signaling to sustain chemoresistance. Front Oncol 2022; 12:906260. [PMID: 36330477 PMCID: PMC9623568 DOI: 10.3389/fonc.2022.906260] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/15/2022] [Indexed: 08/05/2023] Open
Abstract
Chemoresistance in colorectal cancer initiating cells (CICs) involves the sustained activation of multiple drug resistance (MDR) and WNT/β-catenin signaling pathways, as well as of alternatively spliced-isoforms of CD44 containing variable exon-6 (CD44v6). In spite of its importance, mechanisms underlying the sustained activity of WNT/β-catenin signaling have remained elusive. The presence of binding elements of the β-catenin-interacting transcription factor TCF4 in the MDR1 and CD44 promoters suggests that crosstalk between WNT/β-catenin/TCF4-activation and the expression of the CD44v6 isoform mediated by FOLFOX, a first-line chemotherapeutic agent for colorectal cancer, could be a fundamental mechanism of FOLFOX resistance. Our results identify that FOLFOX treatment induced WNT3A secretion, which stimulated a positive feedback loop coupling β-catenin signaling and CD44v6 splicing. In conjunction with FOLFOX induced WNT3A signal, specific CD44v6 variants produced by alternative splicing subsequently enhance the late wave of WNT/β-catenin activation to facilitate cell cycle progression. Moreover, we revealed that FOLFOX-mediated sustained WNT signal requires the formation of a CD44v6-LRP6-signalosome in caveolin microdomains, which leads to increased FOLFOX efflux. FOLFOX-resistance in colorectal CICs occurs in the absence of tumor-suppressor disabled-2 (DAB2), an inhibitor of WNT/β-catenin signaling. Conversely, in sensitive cells, DAB2 inhibition of WNT-signaling requires interaction with a clathrin containing CD44v6-LRP6-signalosome. Furthermore, full-length CD44v6, once internalized through the caveolin-signalosome, is translocated to the nucleus where in complex with TCF4, it binds to β-catenin/TCF4-regulated MDR1, or to CD44 promoters, which leads to FOLFOX-resistance and CD44v6 transcription through transcriptional-reprogramming. These findings provide evidence that targeting CD44v6-mediated LRP6/β-catenin-signaling and drug efflux may represent a novel approach to overcome FOLFOX resistance and inhibit tumor progression in colorectal CICs. Thus, sustained drug resistance in colorectal CICs is mediated by overexpression of CD44v6, which is both a functional biomarker and a therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Shibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| | - Vincent C. Hascall
- Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, OH, United States
| | - Nikos Karamanos
- University of Patras, Matrix Pathobiology Res. Group, Department of Chemistry, Patras, Greece
| | - Roger R. Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| |
Collapse
|
42
|
Wang H, Jin X, Zhang Y, Wang Z, Zhang T, Xu J, Shen J, Zan P, Sun M, Wang C, Hua Y, Ma X, Sun W. Inhibition of sphingolipid metabolism in osteosarcoma protects against CD151-mediated tumorigenicity. Cell Biosci 2022; 12:169. [PMID: 36209197 PMCID: PMC9548188 DOI: 10.1186/s13578-022-00900-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma is the most common primary bone tumor, with a poor prognosis owing to the lack of efficient molecular-based targeted therapies. Previous studies have suggested an association between CD151 and distinct consequences in osteosarcoma tumorigenicity. However, the potential of CD151 as a therapeutic target has not yet been sufficiently explored. Here, we performed integrated transcriptomic and metabolomic analyses of osteosarcoma and identified sphingolipid metabolism as the top CD151-regulated pathway. CD151 regulates sphingolipid metabolism primarily through SPTCL1, the first rate-limiting enzyme in sphingolipid biosynthesis. Mechanistically, depletion of CD151 enhanced c-myc polyubiquitination and subsequent degradation. c-myc is vital for the transcriptional activation of SPTLC1. Functionally, sphingolipid synthesis and the SPTLC1 inhibitor, myriocin, significantly suppressed the clonogenic growth of CD151-overexpression cells. Importantly, myriocin selectively restrained CD151-high expression tumor growth in preclinical patient-derived xenograft models. Collectively, these data establish that CD151 is a key mediator of sphingolipid metabolism and provide a new approach to developing novel CD151-based targeted therapies for osteosarcoma.
Collapse
Affiliation(s)
- Hongsheng Wang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Xinmeng Jin
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Yangfeng Zhang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhuoying Wang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Tao Zhang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Xu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Jiakang Shen
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Pengfei Zan
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Mengxiong Sun
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Chongren Wang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Yingqi Hua
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Xiaojun Ma
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| | - Wei Sun
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080 China ,grid.412478.c0000 0004 1760 4628Shanghai Bone Tumor Institution, Shanghai, China
| |
Collapse
|
43
|
Ramos EK, Tsai CF, Jia Y, Cao Y, Manu M, Taftaf R, Hoffmann AD, El-Shennawy L, Gritsenko MA, Adorno-Cruz V, Schuster EJ, Scholten D, Patel D, Liu X, Patel P, Wray B, Zhang Y, Zhang S, Moore RJ, Mathews JV, Schipma MJ, Liu T, Tokars VL, Cristofanilli M, Shi T, Shen Y, Dashzeveg NK, Liu H. Machine learning-assisted elucidation of CD81-CD44 interactions in promoting cancer stemness and extracellular vesicle integrity. eLife 2022; 11:e82669. [PMID: 36193887 PMCID: PMC9581534 DOI: 10.7554/elife.82669] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
Tumor-initiating cells with reprogramming plasticity or stem-progenitor cell properties (stemness) are thought to be essential for cancer development and metastatic regeneration in many cancers; however, elucidation of the underlying molecular network and pathways remains demanding. Combining machine learning and experimental investigation, here we report CD81, a tetraspanin transmembrane protein known to be enriched in extracellular vesicles (EVs), as a newly identified driver of breast cancer stemness and metastasis. Using protein structure modeling and interface prediction-guided mutagenesis, we demonstrate that membrane CD81 interacts with CD44 through their extracellular regions in promoting tumor cell cluster formation and lung metastasis of triple negative breast cancer (TNBC) in human and mouse models. In-depth global and phosphoproteomic analyses of tumor cells deficient with CD81 or CD44 unveils endocytosis-related pathway alterations, leading to further identification of a quality-keeping role of CD44 and CD81 in EV secretion as well as in EV-associated stemness-promoting function. CD81 is coexpressed along with CD44 in human circulating tumor cells (CTCs) and enriched in clustered CTCs that promote cancer stemness and metastasis, supporting the clinical significance of CD81 in association with patient outcomes. Our study highlights machine learning as a powerful tool in facilitating the molecular understanding of new molecular targets in regulating stemness and metastasis of TNBC.
Collapse
Affiliation(s)
- Erika K Ramos
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National LaboratoryWashingtonUnited States
| | - Yuzhi Jia
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
| | - Yue Cao
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M UniversityCollege StationUnited States
| | - Megan Manu
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
| | - Rokana Taftaf
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Andrew D Hoffmann
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
| | | | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National LaboratoryWashingtonUnited States
| | | | - Emma J Schuster
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - David Scholten
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Dhwani Patel
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
| | - Xia Liu
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Department of Toxicology and Cancer Biology, University of KentuckyLexingtonUnited States
| | - Priyam Patel
- Quantitative Data Science Core, Center for Genetic Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Brian Wray
- Quantitative Data Science Core, Center for Genetic Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Youbin Zhang
- Department of Medicine, Hematology/Oncology Division, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Shanshan Zhang
- Pathology Core Facility, Northwestern UniversityChicagoUnited States
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National LaboratoryWashingtonUnited States
| | - Jeremy V Mathews
- Pathology Core Facility, Northwestern UniversityChicagoUnited States
| | - Matthew J Schipma
- Quantitative Data Science Core, Center for Genetic Medicine, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National LaboratoryWashingtonUnited States
| | - Valerie L Tokars
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
| | - Massimo Cristofanilli
- Department of Medicine, Hematology/Oncology Division, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National LaboratoryWashingtonUnited States
| | - Yang Shen
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M UniversityCollege StationUnited States
| | | | - Huiping Liu
- Department of Pharmacology, Northwestern UniversityChicagoUnited States
- Department of Medicine, Hematology/Oncology Division, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| |
Collapse
|
44
|
Himes BT, Fain CE, Tritz ZP, Nesvick CL, Jin-Lee HJ, Geiger PA, Peterson TE, Jung MY, Parney IF. Use of heparin to rescue immunosuppressive monocyte reprogramming by glioblastoma-derived extracellular vesicles. J Neurosurg 2022; 138:1291-1301. [PMID: 36115048 DOI: 10.3171/2022.6.jns2274] [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: 01/11/2022] [Accepted: 06/17/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The profound immunosuppression found in glioblastoma (GBM) patients is a critical barrier to effective immunotherapy. Multiple mechanisms of tumor-mediated immune suppression exist, and the induction of immunosuppressive monocytes such as myeloid-derived suppressor cells (MDSCs) is increasingly appreciated as a key part of this pathology. GBM-derived extracellular vesicles (EVs) can induce the formation of MDSCs. The authors sought to identify the molecular consequences of these interactions in myeloid cells in order to identify potential targets that could pharmacologically disrupt GBM EV-monocyte interaction as a means to ameliorate tumor-mediated immune suppression. Heparin-sulfate proteoglycans (HSPGs) are a general mechanism by which EVs come into association with their target cells, and soluble heparin has been shown to interfere with EV-HSPG interactions. The authors sought to assess the efficacy of heparin treatment for mitigating the effects of GBM EVs on the formation of MDSCs. METHODS GBM EVs were collected from patient-derived cell line cultures via staged ultracentrifugation and cocultured with monocytes collected from apheresis cones from healthy blood donors. RNA was isolated from EV-conditioned and unconditioned monocytes after 72 hours of coculture, and RNA-sequencing analysis performed. For the heparin treatment studies, soluble heparin was added at the time of EV-monocyte coculture and flow cytometry analysis was performed 72 hours later. After the initial EV-monocyte coculture period, donor-matched T-cell coculture studies were performed by adding fluorescently labeled and stimulated T cells for 5 days of coculture. RESULTS Transcriptomic analysis of GBM EV-treated monocytes demonstrated downregulation of several important immunological and metabolic pathways, with upregulation of the pathways associated with synthesis of cholesterol and HSPG. Heparin treatment inhibited association between GBM EVs and monocytes in a dose-dependent fashion, which resulted in a concomitant reduction in MDSC formation (p < 0.01). The authors further demonstrated that reduced MDSC formation resulted in a partial rescue of immune suppression, as measured by effects on activated donor-matched T cells (p < 0.05). CONCLUSIONS The authors demonstrated that GBM EVs induce broad but reproducible reprogramming in monocytes, with enrichment of pathways that may portend an immunosuppressive phenotype. The authors further demonstrated that GBM EV-monocyte interactions are potentially druggable targets for overcoming tumor-mediated immune suppression, with heparin inhibition of EV-monocyte interactions demonstrating proof of principle.
Collapse
Affiliation(s)
| | - Cori E Fain
- 2Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | - Ian F Parney
- 1Department of Neurologic Surgery and.,2Department of Immunology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
45
|
Zhang J, Shi W, Qu D, Yu T, Qi C, Fu H. Extracellular vesicle therapy for traumatic central nervous system disorders. Stem Cell Res Ther 2022; 13:442. [PMID: 36056445 PMCID: PMC9438220 DOI: 10.1186/s13287-022-03106-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/31/2022] [Indexed: 11/10/2022] Open
Abstract
Traumatic central nervous system (CNS) disorders have catastrophic effects on patients, and, currently, there is no effective clinical treatment. Cell transplantation is a common treatment for traumatic CNS injury in animals. In recent years, an increasing number of studies have reported that the beneficial effect of transplanted cells for CNS repair is mediated primarily through the extracellular vesicles (EVs) secreted by the cells, in which microRNAs play a major role. Accordingly, numerous studies have evaluated the roles and applications of EVs secreted by different cell types in neurological diseases. Furthermore, due to their unique biological features, EVs are used as disease biomarkers and drug delivery systems for disease prevention and treatment. We discuss current knowledge related to EVs, focusing on the mechanism underlying their effects on traumatic CNS diseases, and summarize existing research on the potential clinical utility of EVs as disease biomarkers and drug delivery systems.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.,Medical Department of Qingdao University, Qingdao, China
| | - Weipeng Shi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.,Medical Department of Qingdao University, Qingdao, China
| | - Di Qu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.,Medical Department of Qingdao University, Qingdao, China
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
46
|
Hu Y, Guo J, Jang H, Liu A, Ma L, Ren D, Wang F. Long Non-Coding RNA T Cell Leukemia/Lymphoma 6 Inhibits the Proliferation and Invasion of Breast Cancer Cells by Down-Regulating miR-665. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Breast cancer (BC), which is most commonly seen in women, has become the second most common cause of cancer death in the United States. The number of women dying from BC is increasing every year, especially in the developing countries that fall behind in terms of economy and technologies.
Therefore, it is of great necessity to find potential targets to effectively treat this disease. In this study, RT-qPCR was performed to detect the expressions of TCL6, miR-665, and CD82. CCK-8 and immunofluorescence assays were conducted for the assessment of BC cell proliferation. The invasion
and migration of BC cells were detected by transwell and wound healing assays, respectively. Luciferase reporter assay was used to verify the combination of TCL6 and miR-665, and the binding of miR-665 and CD82. Moreover, the proliferation and migration of related proteins were measured by
western blot. The results showed that TCL6 was low expressed in BC cells, but overexpression of TCL6 inhibited the proliferation, migration, and invasion of BC cells. On the contrary, miR-665 was highly expressed in BC cells, while its expression was negatively correlated with TCL6 as suggested
by RT-qPCR assay. Furthermore, the inhibitory effects of TCL6 overexpression on the proliferation, migration, and invasion of BC cells were reversed by miR-665 mimic. Afterwards, the binding sites between miR-665 and CD82 were verified by luciferase reporter assay. Overexpression of TCL6 increased
the level of CD82 in BC cells, but this effect was reversed by miR-665 mimic as well. In conclusion, the present study has presented the fact that TCL6 could enhance the expression of CD82 by down-regulating the expression of miR-665.
Collapse
Affiliation(s)
- Yaofeng Hu
- Department of Breast Surgery, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Jing Guo
- The Second Group of Geriatrics Department, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Hongnan Jang
- Department of Breast Surgery, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Ailan Liu
- Clinical Laboratory, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Lijun Ma
- Department of Breast Surgery, Shanxi Bethune Hospital, Taiyuan Shanxi 030032, China
| | - Dongliang Ren
- Department of Breast Surgery, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| | - Fusheng Wang
- Department of Breast Surgery, The Second Hospital of Shanxi Medical University, Taiyuan Shanxi 030001, China
| |
Collapse
|
47
|
Regulates CD9 Expression and Dissemination of B Lymphoblasts. Leuk Res 2022; 123:106964. [DOI: 10.1016/j.leukres.2022.106964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022]
|
48
|
Santos MF, Rappa G, Fontana S, Karbanová J, Aalam F, Tai D, Li Z, Pucci M, Alessandro R, Morimoto C, Corbeil D, Lorico A. Anti-Human CD9 Fab Fragment Antibody Blocks the Extracellular Vesicle-Mediated Increase in Malignancy of Colon Cancer Cells. Cells 2022; 11:2474. [PMID: 36010551 PMCID: PMC9406449 DOI: 10.3390/cells11162474] [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: 07/08/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 12/08/2022] Open
Abstract
Intercellular communication between cancer cells themselves or with healthy cells in the tumor microenvironment and/or pre-metastatic sites plays an important role in cancer progression and metastasis. In addition to ligand-receptor signaling complexes, extracellular vesicles (EVs) are emerging as novel mediators of intercellular communication both in tissue homeostasis and in diseases such as cancer. EV-mediated transfer of molecular activities impacting morphological features and cell motility from highly metastatic SW620 cells to non-metastatic SW480 cells is a good in vitro example to illustrate the increased malignancy of colorectal cancer leading to its transformation and aggressive behavior. In an attempt to intercept the intercellular communication promoted by EVs, we recently developed a monovalent Fab fragment antibody directed against human CD9 tetraspanin and showed its effectiveness in blocking the internalization of melanoma cell-derived EVs and the nuclear transfer of their cargo proteins into recipient cells. Here, we employed the SW480/SW620 model to investigate the anti-cancer potential of the anti-CD9 Fab antibody. We first demonstrated that most EVs derived from SW620 cells contain CD9, making them potential targets. We then found that the anti-CD9 Fab antibody, but not the corresponding divalent antibody, prevented internalization of EVs from SW620 cells into SW480 cells, thereby inhibiting their phenotypic transformation, i.e., the change from a mesenchymal-like morphology to a rounded amoeboid-like shape with membrane blebbing, and thus preventing increased cell migration. Intercepting EV-mediated intercellular communication in the tumor niche with an anti-CD9 Fab antibody, combined with direct targeting of cancer cells, could lead to the development of new anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Mark F. Santos
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| | - Germana Rappa
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| | - Simona Fontana
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Jana Karbanová
- Tissue Engineering Laboratories, Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Feryal Aalam
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| | - Derek Tai
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| | - Zhiyin Li
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| | - Marzia Pucci
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Riccardo Alessandro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Denis Corbeil
- Tissue Engineering Laboratories, Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Aurelio Lorico
- Department of Basic Sciences, Touro University College of Medicine, Henderson, NV 89014, USA
| |
Collapse
|
49
|
Punzo F, Argenziano M, Tortora C, Di Paola A, Mutarelli M, Pota E, Di Martino M, Di Pinto D, Marrapodi MM, Roberti D, Rossi F. Effect of CB2 Stimulation on Gene Expression in Pediatric B-Acute Lymphoblastic Leukemia: New Possible Targets. Int J Mol Sci 2022; 23:8651. [PMID: 35955786 PMCID: PMC9369382 DOI: 10.3390/ijms23158651] [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: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 01/27/2023] Open
Abstract
Acute lymphoblastic leukemia type B (B-ALL) is the most common kind of pediatric leukemia, characterized by the clonal proliferation of type B lymphoid stem cells. Important progress in ALL treatments led to improvements in long-term survival; nevertheless, many adverse long-term consequences still concern the medical community. Molecular and cellular target therapies, together with immunotherapy, are promising strategies to overcome these concerns. Cannabinoids, enzymes involved in their metabolism, and cannabinoid receptors type 1 (CB1) and type 2 (CB2) constitute the endocannabinoid system, involved in inflammation, immune response, and cancer. CB2 receptor stimulation exerts anti-proliferative and anti-invasive effects in many tumors. In this study, we evaluated the effects of CB2 stimulation on B-ALL cell lines, SUP-B15, by RNA sequencing, Western blotting, and ELISA. We observe a lower expression of CB2 in SUP-B15 cells compared to lymphocytes from healthy subjects, hypothesizing its involvement in B-ALL pathogenesis. CB2 stimulation reduces the expression of CD9, SEC61G, TBX21, and TMSB4X genes involved in tumor growth and progression, and also negatively affects downstream intracellular pathways. Our findings suggest an antitumor role of CB2 stimulation in B-ALL, and highlight a functional correlation between CB2 receptors and specific anti-tumoral pathways, even though further investigations are needed.
Collapse
Affiliation(s)
- Francesca Punzo
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Maura Argenziano
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Chiara Tortora
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Margherita Mutarelli
- Istituto di Scienze Applicate e Sistemi Intelligenti “Eduardo Caianiello” ISASI-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy;
| | - Elvira Pota
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Martina Di Martino
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Daniela Di Pinto
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Domenico Roberti
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, Via Luigi De Crecchio 4, 80138 Naples, Italy; (F.P.); (M.A.); (C.T.); (A.D.P.); (E.P.); (M.D.M.); (D.D.P.); (M.M.M.); (D.R.)
| |
Collapse
|
50
|
Abstract
Tissue engineering and regenerative medicine (TERM) may be defined as a translational discipline focused on the development of novel techniques, devices, and materials to replace or repair injured or diseased tissue and organs. The main approaches typically use cells, scaffolds, and signaling molecules, either alone or in combination, to promote repair and regeneration. Although cells are required to create new functional tissue, the source of cells, either from an exogenous allogeneic or autologous source or through the recruitment of endogenous (autologous) cells, is technically challenging and risks the host rejection of new tissue. Regardless of the cell source, these approaches also require appropriate instruction for proliferation, differentiation, and in vivo spatial organization to create new functional tissue. Such instruction is supplied through the microenvironment where cells reside, environments which largely consist of the extracellular matrix (ECM). The specific components of the ECM, and broadly the extracellular space, responsible for promoting tissue regeneration and repair, are not fully understood, however extracellular vesicles (EVs) found in body fluids and solid phases of ECM have emerged as key mediators of tissue regeneration and repair. Additionally, these EVs might serve as potential cell-free tools in TERM to promote tissue repair and regeneration with minimal risk for host rejection and adverse sequelae. The past two decades have shown a substantial interest in understanding the therapeutic role of EVs and their applications in the context of TERM. Therefore, the purpose of this review is to highlight the fundamental characteristics of EVs, the current pre-clinical and clinical applications of EVs in TERM, and the future of EV-based strategies in TERM.
Collapse
|