1
|
Jaime-Casas S, Tripathi A, Pal SK, Yip W. Clinical Implications of the Molecular and Genomic Landscape of Upper Tract Urothelial Carcinoma. Curr Urol Rep 2024; 26:11. [PMID: 39379745 PMCID: PMC11461588 DOI: 10.1007/s11934-024-01245-1] [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] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE OF REVIEW Upper tract urothelial carcinoma (UTUC) is an aggressive entity with treatment strategies mirroring bladder cancer. Genomic and molecular profiling allows for a better characterization of this disease and allows for patient-tailored approaches. We aim to describe the genomic and molecular implications of this disease. RECENT FINDINGS Technological advances have the potential for early diagnosis and precise molecular analysis in patients with UTUC. Genomic profile clustering, specific mRNA signatures, and pathway-specific protein abundance tools have oncologic and clinical implications. We describe their utility in the context of this disease. In the era of precision medicine, designing clinical trials that explore the diagnostic and prognostic implications of biomolecular signatures in the context of UTUC is of utmost importance. Promising advances in this arena provide tools for physicians to avoid overtreatment in this patient population.
Collapse
Affiliation(s)
- Salvador Jaime-Casas
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Abhishek Tripathi
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Sumanta K Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Wesley Yip
- Division of Urology and Urologic Oncology, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
2
|
Xiang Z, Yu S, Xu Y, Xiong H, Hu D, Li Q, Wu Z. CH25H Promotes Autophagy and Regulates the Malignant Progression of Laryngeal Squamous Cell Carcinoma Through the PI3K-AKT Pathway. Cancer Med 2024; 13:e70312. [PMID: 39428922 PMCID: PMC11491687 DOI: 10.1002/cam4.70312] [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: 01/09/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Laryngeal squamous cell carcinoma (LSCC) is a type of cancer of the respiratory tract that often presents with subtle symptoms at the early stage and is susceptible to recurrence and metastasis. MATERIALS AND METHODS To find out key regulatory genes involved in LSCC development, we downloaded LSCC-related sequencing datasets for bioinformatics analysis. WGCNA was performed on GSE142083 and differential analysis was conducted on GSE51985 and TCGA-HNSC. Intersection genes were taken from the above three datasets. To confirm the function of genes, we overexpressed and knocked down genes in cells and treated them with autophagy agonist Rapamycin and PI3K-AKT pathway inhibitor. At the cellular level, the expression of CH25H, autophagy-related proteins (LC3 I, LC3 II, p62, and Beclin 1), and PI3K-AKT pathway-related proteins (PI3K, AKT, and p-AKT) were assessed via Western blot; the mRNA level of CH25H was evaluated through qRT-PCR; the cell activity was examined by CCK8; the apoptosis was assessed through flow cytometry; and the cell migration and invasion were assessed through wound healing and Transwell assays. RESULTS Through bioinformatics analysis, we screened 7 genes (CH25H, NELL2, STC2, TMEM158, ZIC2, HOXD11, and HOXD10). Ultimately, CH25H was selected for follow-up experiments. By detecting CH25H expression in human immortalized keratinocytes (HaCaT) and LSCC cells (Tu-686, SNU899, and AMC-HN-8), it was found out that CH25H expression was higher in HaCaT cells than in LSCC cells. To elucidate the role of CH25H in LSCC development, we overexpressed CH25H in Tu-686 cells and downregulated its expression in AMC-HN-8 cells. CH25H was revealed to reduce the proliferation, activity, invasion, and migration of LSCC cells while increasing their apoptosis levels. Significant changes were also observed in the expressions of autophagy- and PI3K-AKT pathway-related proteins. To further investigate the roles of autophagy and the PI3K-AKT pathway in LSCC development, we respectively employed autophagy agonists and inhibitors targeting the PI3K-AKT pathway to intervene the cells, and found that CH25H regulated the PI3K-AKT pathway to promote autophagy, thus enhancing the apoptosis of LSCC cells. We further investigated CH25H's impact on tumor growth, autophagy, and the PI3K-AKT pathway at the animal level and found that CH25H promoted autophagy of LSCC cells and inhibited the PI3K-AKT pathway, and ultimately inhibiting the progression of LSCC. CONCLUSIONS In summary, CH25H promotes autophagy and affects the malignant progression of LSCC through the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Zhenfei Xiang
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Senquan Yu
- Department of OncologyThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yuxin Xu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Huacai Xiong
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Danfei Hu
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Qun Li
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Zhenhua Wu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
3
|
Wang S, Kong H, Zhuo C, Liu L, Lv S, Cheng D, Lao YH, Tao Y, Li M. Functionalized extracellular nanovesicles as advanced CRISPR delivery systems. Biomater Sci 2024; 12:3480-3499. [PMID: 38808607 DOI: 10.1039/d4bm00054d] [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: 05/30/2024]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR) system, an emerging tool for genome editing, has garnered significant public interest for its potential in treating genetic diseases. Despite the rapid advancements in CRISPR technology, the progress in developing effective delivery strategies lags, impeding its clinical application. Extracellular nanovesicles (EVs), either in their endogenous forms or with engineered modifications, have emerged as a promising solution for CRISPR delivery. These EVs offer several advantages, including high biocompatibility, biological permeability, negligible immunogenicity, and straightforward production. Herein, we first summarize various types of functional EVs for CRISPR delivery, such as unmodified, modified, engineered virus-like particles (VLPs), and exosome-liposome hybrid vesicles, and examine their distinct intracellular pathways. Then, we outline the cutting-edge techniques for functionalizing extracellular vesicles, involving producer cell engineering, vesicle engineering, and virus-like particle engineering, emphasizing the diverse CRISPR delivery capabilities of these nanovesicles. Lastly, we address the current challenges and propose rational design strategies for their clinical translation, offering future perspectives on the development of functionalized EVs.
Collapse
Affiliation(s)
- Siqing Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Chenya Zhuo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Li Liu
- Department of Gynecology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Du Cheng
- PCFM Lab of Ministry of Education, School of Material Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
4
|
Li L, Jiang H, Zeng B, Wang X, Bao Y, Chen C, Ma L, Yuan J. Liquid biopsy in lung cancer. Clin Chim Acta 2024; 554:117757. [PMID: 38184141 DOI: 10.1016/j.cca.2023.117757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Lung cancer is a highly prevalent malignancy worldwide and the primary cause of mortality. The absence of systematic and standardized diagnostic approaches for identifying potential pulmonary nodules, early-stage cancers, and indeterminate tumors has led clinicians to consider tissue biopsy and pathological sections as the preferred method for clinical diagnosis, often regarded as the gold standard. The conventional tissue biopsy is an invasive procedure that does not adequately capture the diverse characteristics and evolving nature of tumors. Recently, the concept of 'liquid biopsy' has gained considerable attention as a promising solution. Liquid biopsy is a non-invasive approach that facilitates repeated analysis, enabling real-time monitoring of tumor recurrence, metastasis, and response to treatment. Currently, liquid biopsy includes circulating tumor cells, circulating cell-free DNA, circulating tumor DNA, circulating cell-free RNA, extracellular vesicles, and other proteins and metabolites. With rapid progress in molecular technology, liquid biopsy has emerged as a highly promising and intriguing approach, yielding compelling results. This article critically examines the significant role and potential clinical implications of liquid biopsy in the diagnosis, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Lan Li
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haixia Jiang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Bingjie Zeng
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Xianzhao Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Yunxia Bao
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Changqiang Chen
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Jin Yuan
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
5
|
Li Y, Fan L, Yan A, Ren X, Zhao Y, Hua B. Exosomal miR-361-3p promotes the viability of breast cancer cells by targeting ETV7 and BATF2 to upregulate the PAI-1/ERK pathway. J Transl Med 2024; 22:112. [PMID: 38282047 PMCID: PMC10823750 DOI: 10.1186/s12967-024-04914-4] [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: 09/08/2023] [Accepted: 01/21/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Malignant progression is the major cause of poor prognosis in breast cancer (BC) patients. Plasma exosomal miRNAs have been reported to be involved in tumor progression, but their roles in BC remain unclear. METHODS We performed plasma exosomal miRNA sequencing on 45 individuals, including healthy controls and nonmetastatic and metastatic BC patients. We examined the correlation between miRNA expression in tumor tissues and plasma exosomes in BC patients by qRT‒PCR. The effects of exosomal miR-361-3p on BC cells were determined by CellTiter-Glo, migration and wound healing assays. The target genes of miR-361-3p and downstream pathways were explored by dual-luciferase reporter assay, RNA knockdown, rescue experiments, and western blotting. We utilized murine xenograft model to further assess the impact of plasma exosomal miR-361-3p on the malignant progression of BC. RESULTS We found that the expression level of plasma exosomal miR-361-3p gradually increased with malignant progression in BC patients, and the expression of miR-361-3p in plasma exosomes and BC tissues was positively correlated. Consistently, exosomal miR-361-3p enhanced the migration and proliferation of two BC cell lines, MDA-MB-231 and SK-BR-3. Furthermore, our data showed that miR-361-3p inhibited two novel target genes, ETV7 and BATF2, to activate the PAI-1/ERK pathway, leading to increased BC cell viability. Finally, the consistency of the in vivo experimental results supported that elevated plasma exosomal miR-361-3p promote the malignant progression of BC. CONCLUSIONS We found for the first time that plasma exosomal miR-361-3p was associated with malignant progression in BC patients. Mechanistically, exosomal miR-361-3p can enhance the migration and proliferation of BC cells by targeting the ETV7 and BATF2/PAI-1/ERK pathways. Our data suggest that plasma exosomal miR-361-3p has the potential to serve as a biomarker for predicting malignant progression in BC patients.
Collapse
Affiliation(s)
- Yao Li
- Breast center, Department of Thyroid-Breast-Hernia Surgery, Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Lei Fan
- Breast center, Department of Thyroid-Breast-Hernia Surgery, Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - An Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, People's Republic of China
| | - Xiaotian Ren
- Breast center, Department of Thyroid-Breast-Hernia Surgery, Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yanyang Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, People's Republic of China
| | - Bin Hua
- Breast center, Department of Thyroid-Breast-Hernia Surgery, Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Yang YC, Jiang Q, Yang KP, Wang L, Sethi G, Ma Z. Extracellular vesicle-mediated ferroptosis, pyroptosis, and necroptosis: potential clinical applications in cancer therapy. Cell Death Discov 2024; 10:23. [PMID: 38216595 PMCID: PMC10786909 DOI: 10.1038/s41420-024-01799-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 12/15/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024] Open
Abstract
Extracellular vesicles (EVs) have gained increasing recognition as significant regulators of intercellular communication in various physiological and pathological processes. These vesicles play a pivotal role in cancer progression by facilitating the transfer of diverse cargoes, including lipids, proteins, and nucleic acids. Regulated cell death (RCD), the orderly and autonomous death of cells, is controlled by a variety of biomacromolecules and, in turn, influences various biological processes and cancer progression. Recent studies have demonstrated that EV cargoes regulate diverse oncogenes and tumor suppressors to mediate different nonapoptotic forms of RCD, notably ferroptosis, pyroptosis, and necroptosis. Nevertheless, comprehensive exploration of EV-mediated nonapoptotic RCD forms in the context of cancer has not been performed. This review summarizes the progress regarding the biological functions and underlying mechanisms of EVs in mediating nonapoptotic RCD by delivery of cargoes to regulate tumor progression. Additionally, the review delves into the potential clinical applications of EV-mediated cell death and its significance in the areas of cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yi-Chi Yang
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, 434023, Jingzhou, Hubei, China
| | - Qian Jiang
- Honghu Hospital of Traditional Chinese Medicine, 433200, Honghu, China
- Digestive Disease Research Institution of Yangtze University, Yangtze University, 434023, Jingzhou, China
| | - Ke-Ping Yang
- Department of Cardiology, Jingzhou Hospital Affiliated to Yangtze University, 434023, Jingzhou, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, 434023, Jingzhou, Hubei, China.
| |
Collapse
|
7
|
Nguyen C, Saint-Pol J, Dib S, Pot C, Gosselet F. 25-Hydroxycholesterol in health and diseases. J Lipid Res 2024; 65:100486. [PMID: 38104944 PMCID: PMC10823077 DOI: 10.1016/j.jlr.2023.100486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Cholesterol is an essential structural component of all membranes of mammalian cells where it plays a fundamental role not only in cellular architecture, but also, for example, in signaling pathway transduction, endocytosis process, receptor functioning and recycling, or cytoskeleton remodeling. Consequently, intracellular cholesterol concentrations are tightly regulated by complex processes, including cholesterol synthesis, uptake from circulating lipoproteins, lipid transfer to these lipoproteins, esterification, and metabolization into oxysterols that are intermediates for bile acids. Oxysterols have been considered for long time as sterol waste products, but a large body of evidence has clearly demonstrated that they play key roles in central nervous system functioning, immune cell response, cell death, or migration and are involved in age-related diseases, cancers, autoimmunity, or neurological disorders. Among all the existing oxysterols, this review summarizes basic as well as recent knowledge on 25-hydroxycholesterol which is mainly produced during inflammatory or infectious situations and that in turn contributes to immune response, central nervous system disorders, atherosclerosis, macular degeneration, or cancer development. Effects of its metabolite 7α,25-dihydroxycholesterol are also presented and discussed.
Collapse
Affiliation(s)
- Cindy Nguyen
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Julien Saint-Pol
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Shiraz Dib
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France
| | - Caroline Pot
- Department of Clinical Neurosciences, Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabien Gosselet
- UR 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Univ. Artois, Lens, France.
| |
Collapse
|
8
|
Oregel-Cortez MI, Frayde-Gómez H, Quintana-González G, García-González V, Vazquez-Jimenez JG, Galindo-Hernández O. Resistin Induces Migration and Invasion in PC3 Prostate Cancer Cells: Role of Extracellular Vesicles. Life (Basel) 2023; 13:2321. [PMID: 38137922 PMCID: PMC10744490 DOI: 10.3390/life13122321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 12/24/2023] Open
Abstract
Resistin is an adipokine with metabolic and inflammatory functions. Epidemiological and translational studies report that an increase in plasma levels and tissue expression of resistin increases the aggressiveness of prostate tumor cells. Extracellular vesicles (EVs) are secreted constitutively and induced by cytokines, growth factors, and calcium and are found in multiple biological fluids such as saliva, serum, semen, and urine. In particular, EVs have been shown to promote tumor progression through the induction of proliferation, growth, angiogenesis, resistance to chemotherapy, and metastasis. However, the role of resistin in the migration, invasion, and secretion of EVs in invasive prostate tumor cells remains to be studied. In the present study, we demonstrate that resistin induces increased migration and invasion in PC3 cells. In addition, these phenomena are accompanied by increased p-FAK levels and increased secretion of MMP-2 and MMP-9 in resistin-treated PC3 cells. Interestingly, EVs isolated from supernatants of PC3 cells treated with resistin induce an increase in migration and invasion accompanied by high MMP-2 and MMP-9 secretion in an autocrine stimulation model. In summary, our data for the first time demonstrate that resistin induces migration and invasion, partly through the secretion of EVs with pro-invasive characteristics in PC3 cells.
Collapse
Affiliation(s)
- Mario Israel Oregel-Cortez
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
- Facultad de Deportes, Universidad Autónoma de Baja California, Mexicali 21289, Baja California, Mexico
| | - Héctor Frayde-Gómez
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
- Hospital Regional de Especialidad No. 30, Instituto Mexicano del Seguro Social, Mexicali 21100, Baja California, Mexico
| | - Georgina Quintana-González
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| | - Victor García-González
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| | - Jose Gustavo Vazquez-Jimenez
- Laboratorio de Fisiología, Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico;
| | - Octavio Galindo-Hernández
- Departamento de Bioquimíca, Facultad de Medicina, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico; (M.I.O.-C.); (H.F.-G.); (G.Q.-G.); (V.G.-G.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21100, Baja California, Mexico
| |
Collapse
|
9
|
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: 2.0] [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
|
10
|
Wu T, Tian Q, Liu R, Xu K, Shi S, Zhang X, Gao L, Yin X, Xu S, Wang P. Inhibitory role of bone marrow mesenchymal stem cells-derived exosome in non-small-cell lung cancer: microRNA-30b-5p, EZH2 and PI3K/AKT pathway. J Cell Mol Med 2023; 27:3526-3538. [PMID: 37698037 PMCID: PMC10660609 DOI: 10.1111/jcmm.17933] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023] Open
Abstract
Exosomal microRNA (miRNA) exerts potential roles in non-small-cell lung cancer (NSCLC). The current study elucidated the role of miR-30b-5p shuttled by bone marrow mesenchymal stem cells (BMSCs)-derived exosomes in treating NSCLC. Bioinformatics analysis was performed with NSCLC-related miRNA microarray GSE169587 and mRNA data GSE74706 obtained for collection of the differentially expressed miRNAs and mRNAs. The relationship between miR-30b-5p and EZH2 was predicted and confirmed. Exosomes were isolated from BMSCs and identified. BMSCs-derived exosomes overexpressing miR-30b-5p were used to establish subcutaneous tumorigenesis models to study the effects of miR-30b-5p, EZH2 and PI3K/AKT signalling pathway on tumour growth. A total of 86 BMSC-exo-miRNAs were differentially expressed in NSCLC. Bioinfomatics analysis found that BMSC-exo-miR-30b-5p could regulate NSCLC progression by targeting EZH2, which was verified by in vitro cell experiments. Besides, the target genes of miR-30b-5p were enriched in PI3K/AKT signalling pathway. Animal experiments validated that BMSC-exo-miR-30b-5p promoted NSCLC cell apoptosis and prevented tumorigenesis in nude mice via EZH2/PI3K/AKT axis. Collectively, the inhibitory role of BMSC-derived exosomes-loaded miR-30b-5p in NSCLC was achieved through blocking the EZH2/PI3K/AKT axis.
Collapse
Affiliation(s)
- Tong Wu
- Graduate School of Zunyi Medical UniversityZunyiChina
| | - Qi Tian
- Department of Pulmonary and Critical Care MedicineThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Ruiji Liu
- Department of Pulmonary and Critical Care MedicineThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Ke Xu
- Graduate School of Hebei Medical UniversityShijiazhuangChina
| | - Shanshan Shi
- Department of Pulmonary and Critical Care MedicineThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Xiudi Zhang
- Graduate School of Hebei Medical UniversityShijiazhuangChina
| | - Liming Gao
- Oncology DepartmentThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Xiaobo Yin
- Department of Pulmonary and Critical Care MedicineThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Shufeng Xu
- Department of Pulmonary and Critical Care MedicineThe First Hospital of QinhuangdaoQinhuangdaoChina
| | - Ping Wang
- Department of Pulmonary and Critical Care MedicineChinese People's Liberation Army General HospitalBeijingChina
| |
Collapse
|
11
|
Liu W, Jin M, Chen Q, Li Q, Xing X, Luo Y, Sun X. Insight into extracellular vesicles in vascular diseases: intercellular communication role and clinical application potential. Cell Commun Signal 2023; 21:310. [PMID: 37907962 PMCID: PMC10617214 DOI: 10.1186/s12964-023-01304-z] [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: 07/10/2023] [Accepted: 09/02/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Cells have been increasingly known to release extracellular vesicles (EVs) to the extracellular environment under physiological and pathological conditions. A plethora of studies have revealed that EVs contain cell-derived biomolecules and are found in circulation, thereby implicating them in molecular trafficking between cells. Furthermore, EVs have an effect on physiological function and disease development and serve as disease biomarkers. MAIN BODY Given the close association between EV circulation and vascular disease, this review aims to provide a brief introduction to EVs, with a specific focus on the EV cargoes participating in pathological mechanisms, diagnosis, engineering, and clinical potential, to highlight the emerging evidence suggesting promising targets in vascular diseases. Despite the expansion of research in this field, some noticeable limitations remain for clinical translational research. CONCLUSION This review makes a novel contribution to a summary of recent advances and a perspective on the future of EVs in vascular diseases. Video Abstract.
Collapse
Affiliation(s)
- Wenxiu Liu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Qiuyan Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Xiaoyan Xing
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China.
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
| |
Collapse
|
12
|
Koni M, Lopatina T, Grange C, Sarcinella A, Cedrino M, Bruno S, Buffolo F, Femminò S, Camussi G, Brizzi MF. Circulating extracellular vesicles derived from tumor endothelial cells hijack the local and systemic anti-tumor immune response: Role of mTOR/G-CSF pathway. Pharmacol Res 2023; 195:106871. [PMID: 37506784 DOI: 10.1016/j.phrs.2023.106871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Circulating tumour-derived extracellular vesicles are supposed to contribute to the spreading of distant metastasis. In this study, we investigated the impact of circulating extracellular vesicles derived from tumour-endothelial cells (TEVs) in the expansion of the metastatic bulk. We focus on the role of immune cells in controlling this process using the 4T1 triple negative breast cancer (TNBC) syngeneic model. 4T1 cells were intravenously injected and exposed to circulating TEVs from day 7. The lung, spleen, and bone marrow (BM) were recovered and analysed. We demonstrated that circulating TEVs boost lung metastasis and angiogenesis. FACS and immunohistochemically analyses revealed a significant enrichment of Ly6G+/F4/80+/CD11b+ cells and Ly6G+/F4/80-/CD11b+ in the lung and in the spleen, while Ly6G+/F4/80-/CD11b+ in the BM, indicating the occurrence of a systemic and local immune suppression. TEV immune suppressive properties were further supported by the increased expression of PD-L1, PD-1, and iNOS in the tumour mass. In addition, in vitro experiments demonstrated an increase of CD11+ cells, PD-L1+ myeloid and cancer cells, upregulation of LAG3, CTLA4 and PD-1 in T-cells, release of ROS and NOS, and impaired T-cell-mediated cytotoxic effect in co-culture of TEVs-preconditioned PBMCs and cancer cells. Granulocyte-colony stimulating factor (G-CSF) level was increased in vivo, and was involved in reshaping the immune response. Mechanistically, we also found that mTOR enriched TEVs support G-CSF release and trigger the phosphorylation of the S6 (Ser235/236) mTOR downstream target. Overall, we provided evidence that circulating TEVs enriched in mTOR supported G-CSF release thereby granting tumour immune suppression and metastasis outgrowth.
Collapse
Affiliation(s)
- Malvina Koni
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tatiana Lopatina
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabrizio Buffolo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | |
Collapse
|
13
|
Fan L, Xu G, Zeng X. M2 macrophage-derived extracellular vesicles augment immune evasion and development of colorectal cancer via a circRNA_CCDC66/microRNA-342-3p/metadherin axis. Cytotechnology 2023; 75:293-308. [PMID: 37389129 PMCID: PMC10299985 DOI: 10.1007/s10616-023-00577-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] [Received: 11/17/2022] [Accepted: 03/25/2023] [Indexed: 07/01/2023] Open
Abstract
The M2 macrophages are major components in the tumor microenvironment and are closely linked to immune suppression and tumor metastasis. This work focuses on how M2 macrophage-derived extracellular vesicles (EVs) affect colorectal cancer (CRC) progression. THP-1 monocytes were induced to differentiate to M0 or M2 macrophages, and the macrophage-derived EVs (M0-EVs and M2-EVs, respectively) were collected and identified. The M2-EVs stimulation augmented proliferation, mobility, and the in vivo tumorigenic activity of CRC cells. Circular RNA_CCDC66 (circ_CCDC66) was highly enriched in M2-EVs and could be delivered into CRC cells. The RNA pull-down and luciferase assays showed that circ_CCDC66 could competitively bind to microRNA (miR)-342-3p, therefore restoring the expression of metadherin (MTDH) mRNA, a target transcript of miR-342-3p. Suppression of circ_CCDC66 in the M2-EVs or specific knockdown of MTDH in CRC significantly blocked the growth and mobility of CRC cells. However, miR-342-3p inhibition restored the malignant phenotype of cancer cells. Moreover, the MTDH knockdown was found to increase the cytotoxicity of CD8+ T and reduce the protein level of the immune checkpoint PDL1 in CRC cells. In summary, this study reveals that the M2-EVs augment immune evasion and development of CRC by delivering circ_CCDC66 and restoring the MTDH level.
Collapse
Affiliation(s)
- Linfeng Fan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Gannan Medical College, No. 128, Jinling Road, Economic Development Zone, Ganzhou, 341000 Jiangxi People’s Republic of China
| | - Guofeng Xu
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, 341000 Jiangxi People’s Republic of China
| | - Xiangfu Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Gannan Medical College, No. 128, Jinling Road, Economic Development Zone, Ganzhou, 341000 Jiangxi People’s Republic of China
| |
Collapse
|
14
|
Wang Y, Zhong X, He X, Hu Z, Huang H, Chen J, Chen K, Zhao S, Wei P, Li D. Liver metastasis from colorectal cancer: pathogenetic development, immune landscape of the tumour microenvironment and therapeutic approaches. J Exp Clin Cancer Res 2023; 42:177. [PMID: 37480104 PMCID: PMC10362774 DOI: 10.1186/s13046-023-02729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/05/2023] [Indexed: 07/23/2023] Open
Abstract
Colorectal cancer liver metastasis (CRLM) is one of the leading causes of death among patients with colorectal cancer (CRC). Although immunotherapy has demonstrated encouraging outcomes in CRC, its benefits are minimal in CRLM. The complex immune landscape of the hepatic tumour microenvironment is essential for the development of a premetastatic niche and for the colonisation and metastasis of CRC cells; thus, an in-depth understanding of these mechanisms can provide effective immunotherapeutic targets for CRLM. This review summarises recent studies on the immune landscape of the tumour microenvironment of CRLM and highlights therapeutic prospects for targeting the suppressive immune microenvironment of CRLM.
Collapse
Affiliation(s)
- Yaxian Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinyang Zhong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuefeng He
- ZJU-UCLA Joint Center for Medical Education and Research, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijuan Hu
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Huixia Huang
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Jiayu Chen
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Keji Chen
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ping Wei
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.
- Institute of Pathology, Fudan University, Shanghai, China.
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Wang J, Liu Y, Liu F, Gan S, Roy S, Hasan I, Zhang B, Guo B. Emerging extracellular vesicle-based carriers for glioblastoma diagnosis and therapy. NANOSCALE 2023. [PMID: 37337814 DOI: 10.1039/d3nr01667f] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Glioblastoma (GBM) treatment is still a big clinical challenge because of its highly malignant, invasive, and lethal characteristics. After treatment with the conventional therapeutic paradigm of surgery combined with radio- and chemotherapy, patients bearing GBMs generally exhibit a poor prognosis, with high mortality and a high disability rate. The main reason is the existence of the formidable blood-brain barrier (BBB), aggressive growth, and the infiltration nature of GBMs. Especially, the BBB suppresses the delivery of imaging and therapeutic agents to lesion sites, and thus this leads to difficulties in achieving a timely diagnosis and treatment. Recent studies have demonstrated that extracellular vesicles (EVs) exhibit favorable merits including good biocompatibility, a strong drug loading capacity, long circulation time, good BBB crossing efficiency, specific targeting to lesion sites, and high efficiency in the delivery of a variety of cargos for GBM therapy. Importantly, EVs inherit physiological and pathological molecules from the source cells, which are ideal biomarkers for molecularly tracking the malignant progression of GBMs. Herein, we start by introducing the pathophysiology and physiology of GBMs, followed by presenting the biological functions of EVs in GBMs with a special focus on their role as biomarkers for GBM diagnosis and as messengers in the modulation of the GBM microenvironment. Furthermore, we provide an update on the recent progress of using EVs in biology, functionality, and isolation applications. More importantly, we systematically summarize the most recent advances of EV-based carriers for GBM therapy by delivering different drugs including gene/RNA-based drugs, chemotherapy drugs, imaging agents, and combinatory drugs. Lastly, we point out the challenges and prospects of future research on EVs for diagnosing and treating GBMs. We hope this review will stimulate interest from researchers with different backgrounds and expedite the progress of GBM treatment paradigms.
Collapse
Affiliation(s)
- Jingjing Wang
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yue Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Fengbo Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shaoyan Gan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shubham Roy
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ikram Hasan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Bing Guo
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
16
|
Wei J, Ou Z, Tong B, Liao Z, Yang C. Engineered extracellular vesicles as therapeutics of degenerative orthopedic diseases. Front Bioeng Biotechnol 2023; 11:1162263. [PMID: 37362216 PMCID: PMC10289007 DOI: 10.3389/fbioe.2023.1162263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Degenerative orthopedic diseases, as a global public health problem, have made serious negative impact on patients' quality of life and socio-economic burden. Traditional treatments, including chemical drugs and surgical treatments, have obvious side effects and unsatisfactory efficacy. Therefore, biological therapy has become the focus of researches on degenerative orthopedic diseases. Extracellular vesicles (EVs), with superior properties of immunoregulatory, growth support, and drug delivery capabilities, have emerged as a new cell-free strategy for the treatment of many diseases, including degenerative orthopedic diseases. An increasing number of studies have shown that EVs can be engineered through cargo loading, surface modification, and chemical synthesis to improve efficiency, specificity, and safety. Herein, a comprehensive overview of recent advances in engineering strategies and applications of engineered EVs as well as related researches in degenerative orthopedic diseases, including osteoarthritis (OA), osteoporosis (OP), intervertebral disc degeneration (IDD) and osteonecrosis of the femoral head (ONFH), is provided. In addition, we analyze the potential and challenges of applying engineered EVs to clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Cao Yang
- *Correspondence: Zhiwei Liao, ; Cao Yang,
| |
Collapse
|
17
|
Spada S. Methodologies to evaluate the radiation-induced changes on extracellular vesicles. Methods Cell Biol 2023; 180:39-48. [PMID: 37890931 DOI: 10.1016/bs.mcb.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
The extracellular vesicles (EVs) are carriers that actively transfer functional biomolecules between cells affecting the intercellular communication (Pitt, Kroemer, & Zitvogel, 2016). EV signaling has consequences on the targeted cell behavior impacting multiple processes from health to disease, including cancer (Yates et al., 2022). Radiation treatment (RT) is one of the gold standard and effective treatments for cancer, as curative or palliative (Chandra, Keane, Voncken, & Thomas, 2021). RT induces different release of EVs and their cargo is altered. In addition, the uptake of EVs secreted by irradiated cells is affected. Hence, a deep investigation is required to better understand how RT influence the cell-to-cell communication thought signals shuttle by EVs. Here, detailed methods to study the RT effects on EV size and secretion, EV protein expression, EV uptake will be described. Alterations and adaptions might make the protocols applicable to different cell lines, and with different types of RT and dose exposures.
Collapse
Affiliation(s)
- Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
18
|
Mo Y, Liang Z, Lan L, Xiong X, Zhang C, Liu W, Huang H, Fan J, Yang L. Extracellular vesicles derived from cervical cancer cells carrying MCM3AP-AS1 promote angiogenesis and tumor growth in cervical cancer via the miR-93/p21 axis. Exp Cell Res 2023; 428:113621. [PMID: 37137462 DOI: 10.1016/j.yexcr.2023.113621] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/05/2023]
Abstract
Tumor cells can promote angiogenesis by secreting extracellular vesicles (EVs). Meanwhile, tumor-derived EVs can carry long non-coding RNAs to activate pro-angiogenic signaling in endothelial cells. Here, we investigated the role of long non-coding RNA MCM3AP-AS1 carried by cervical cancer (CC) cell-derived EVs in the angiogenesis and the resultant tumor growth in CC, as well as the potential molecular mechanisms. LncRNAs significantly expressed in CC cell-derived EVs and CC were screened, followed by prediction of downstream target genes. EVs were isolated from HcerEpic and CaSki cell supernatants, followed by identification. The expression of MCM3AP-AS1 in CC was analyzed and its interaction with miR-93-p21 was confirmed. Following co-culture system, the role of MCM3AP-AS1 carried by EVs in HUVEC angiogenic ability, CC cell invasion and migration in vitro along with angiogenesis and tumorigenicity in vivo was assayed. MCM3AP-AS1 was overexpressed in CC cell-derived EVs as well as in CC tissues and cell lines. Cervical cancer cell-derived EVs could transfer MCM3AP-AS1 into HUVECs where MCM3AP-AS1 competitively bound to miR-93 and upregulate the expression of the miR-93 target p21 gene. Thus, MCM3AP-AS1 promoted angiogenesis of HUVECs. In the similar manner, MCM3AP-AS1 enhanced CC cell malignant properties. In nude mice, EVs-MCM3AP-AS1 induced angiogenesis and tumor growth. Overall, this study reveals that CC cell-derived EVs may transport MCM3AP-AS1 to promote angiogenesis and tumor growth in CC.
Collapse
Affiliation(s)
- Yuzhen Mo
- Department of Radiotherapy, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China.
| | - Zhishan Liang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530016, China
| | - Liu Lan
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Cici Zhang
- Department of Radiology, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Wei Liu
- Department of Breast Surgery, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Haowei Huang
- Department of Radiotherapy, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Jiangxia Fan
- Department of Radiotherapy, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Li Yang
- Department of Radiotherapy, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| |
Collapse
|
19
|
Li Q, Wang M, Liu L. The role of exosomes in the stemness maintenance and progression of acute myeloid leukemia. Biochem Pharmacol 2023; 212:115539. [PMID: 37024061 DOI: 10.1016/j.bcp.2023.115539] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy of myeloid hematopoietic cells, which is characterized by the aberrant clonal proliferation of immature myeloblasts and compromised hematopoiesis. The leukemic cell population is strongly heterogeneous. Leukemic stem cells (LSCs) are an important leukemic cell subset with stemness characteristics and self-renewal ability, which contribute to the development of refractory or relapsed AML. It is now acknowledged that LSCs develop from hematopoietic stem cells (HSCs) or phenotypically directed cell populations with transcriptional stemness characteristics under selective pressure from the bone marrow (BM) niche. Exosomes are extracellular vesicles containing bioactive substances involved in intercellular communication and material exchange under steady state and pathological conditions. Several studies have reported that exosomes mediate molecular crosstalk between LSCs, leukemic blasts, and stromal cells in the BM niche, promoting LSC maintenance and AML progression. This review briefly describes the process of LSC transformation and the biogenesis of exosomes, highlighting the role of leukemic-cell- and BM-niche-derived exosomes in the maintenance of LSCs and AML progression. In addition, we discuss the potential application of exosomes in the clinic as biomarkers, therapeutic targets, and carriers for targeted drug delivery.
Collapse
Affiliation(s)
- Qian Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mengyuan Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
20
|
Ding L, Li ZL, Zhou Y, Liu NC, Liu SS, Zhang XJ, Liu CC, Zhang DJ, Wang GH, Ma RX. Loss of Sirt1 promotes exosome secretion from podocytes by inhibiting lysosomal acidification in diabetic nephropathy. Mol Cell Endocrinol 2023; 568-569:111913. [PMID: 36990198 DOI: 10.1016/j.mce.2023.111913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/09/2023] [Accepted: 03/12/2023] [Indexed: 03/29/2023]
Abstract
Podocyte injury is a characteristic feature of diabetic nephropathy (DN). The secretion of exosomes in podocytes increases significantly in DN; however, the precise mechanisms remain poorly understood. Here, we demonstrated that Sirtuin1 (Sirt1) was significantly downregulated in podocytes in DN, which correlated negatively with increased exosome secretion. Similar results were observed in vitro. We found that lysosomal acidification in podocytes following high glucose administration was markedly inhibited, resulting in the decreased lysosomal degradation of multivesicular bodies. Mechanistically, we indicated that loss of Sirt1 contributed to the inhibited lysosomal acidification by decreasing the expression of the A subunit of the lysosomal vacuolar-type H+ ATPase proton pump (ATP6V1A) in podocytes. Overexpression of Sirt1 significantly improved lysosomal acidification with increased expression of ATP6V1A and inhibited exosome secretion. These findings suggest that dysfunctional Sirt1-mediated lysosomal acidification is the exact mechanism of increased secretion of exosomes in podocytes in DN, providing insights into potential therapeutic strategies for preventing DN progression.
Collapse
Affiliation(s)
- Lin Ding
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| | - Yan Zhou
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Nan-Chi Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shan-Shan Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xing-Jian Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cong-Cong Liu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Dong-Jie Zhang
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Gui-Hua Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Rui-Xia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
21
|
Wang G, Luo G, Zhao M, Miao H. Significance of exosomes in hepatocellular carcinoma. Front Oncol 2022; 12:1056379. [PMID: 36531059 PMCID: PMC9748478 DOI: 10.3389/fonc.2022.1056379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 09/28/2023] Open
Abstract
Among the most prevalent cancers in the world, hepatocellular carcinoma (HCC) has a high mortality rate. The diagnosis and management of HCC are presently hindered by difficulties in early detection and suboptimal treatment outcomes. Exosomes have been shown to play an important role in hepatocarcinogenesis and can also be used for diagnosis and treatment. In this review, we discussed the research progress on exosomes in hepatocarcinogenesis development, tumor microenvironment remodeling, treatment resistance, and immunosuppression. HCC can be diagnosed and treated by understanding the pathogenesis and identifying early diagnostic markers. This review will be a significant reference for scholars with an initial understanding of the field to fully understand the role of exosomes in the organism.
Collapse
Affiliation(s)
- GuoYun Wang
- Department of Hepatobiliary Surgery, The Second Hospital of Guangdong Medical University, Zhanjiang, China
| | - GaiXiang Luo
- The First Clinical Medical College of Lanzhou University, Gansu Provincial People’s Hospital, Lanzhou, China
| | - MeiJing Zhao
- Department of Hepatobiliary Surgery, The Second Hospital of Guangdong Medical University, Zhanjiang, China
| | - HuiLai Miao
- Department of Hepatobiliary Surgery, The Second Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Liver Injury Diagnosis and Repair, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Xu Y, Luan G, Li Z, Liu Z, Qin G, Chu Y. Tumour-derived exosomal lncRNA SNHG16 induces telocytes to promote metastasis of hepatocellular carcinoma via the miR-942-3p/MMP9 axis. Cell Oncol (Dordr) 2022; 46:251-264. [PMID: 36434360 DOI: 10.1007/s13402-022-00746-w] [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] [Accepted: 11/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) cell-derived exosomal LncRNA SNHG16 is highly expressed and associated with poor overall survival of patients. Telocytes (TCs), as novel interstitial cells, have been reported to promote HCC metastasis. Therefore, in our study, we investigated whether a molecular interaction occurred between exosomal LncSNHG16 and TCs in the tumor microenvironment. METHODS LncSNHG16 expression in HCC tissues and cell lines was measured, and bioinformatics analysis was performed. Exosomes were isolated and purified from HCC cells with LncSNHG16 overexpression/knockdown vectors and cocultured with TCs. Then, markers of the LncSNHG16/miR-942-3p/MMP9 axis were tested in TCs. Transwell assays and cell wound healing assays were designed to examine the invasion and migration of HCC cells after coincubation with TCs. RNA immunoprecipitation (RIP) assays and dual-luciferase gene reporter assays were performed to verify the binding effect of LncSNHG16, miR-942-3p, and MMP9 mRNA. In vivo, experimental animal models were established to confirm the effect of exosomal LncSNHG16-induced MMP9 expression on HCC metastasis. RESULTS Exosomal LncSNHG16 was phagocytized by TCs and downregulated miR-942-3p, which induced targeted MMP9 upregulation, and it had specific binding sites with miR-942-3p in TCs to facilitate the migration of HCC cells in vitro and in vivo. Exosomal LncSNHG16 was found to act as a competing endogenous RNA of the miR-942-3p/MMP9 axis in TCs. CONCLUSION Tumour-derived exosomal LncSNHG16 modulates MMP9 via competitively binding to miR-942-3p in TCs, thus promoting the metastasis of HCC.
Collapse
Affiliation(s)
- Ying Xu
- Shandong Cancer Hospital and Institute, Shandong Fist Medical University and Shandong Academy of Medical Science, No 440, Jiyan Road, Ji'nan, Shandong, China.
| | | | - Zhongchao Li
- Shandong Cancer Hospital and Institute, Shandong Fist Medical University and Shandong Academy of Medical Science, No 440, Jiyan Road, Ji'nan, Shandong, China
| | - Ziming Liu
- Shandong Fist Medical University and Shandong Academy of Medical Science, Ji'nan, Shandong, China
| | - Guangyang Qin
- Shandong Fist Medical University and Shandong Academy of Medical Science, Ji'nan, Shandong, China
| | - Yifu Chu
- Shandong Fist Medical University and Shandong Academy of Medical Science, Ji'nan, Shandong, China
| |
Collapse
|
23
|
Vikramdeo KS, Anand S, Khan MA, Khushman M, Heslin MJ, Singh S, Singh AP, Dasgupta S. Detection of mitochondrial DNA mutations in circulating mitochondria-originated extracellular vesicles for potential diagnostic applications in pancreatic adenocarcinoma. Sci Rep 2022; 12:18455. [PMID: 36323735 PMCID: PMC9630429 DOI: 10.1038/s41598-022-22006-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022] Open
Abstract
There is a complete lack of highly sensitive and specific biomarkers for early pancreatic ductal adenocarcinoma (PDAC) diagnosis, limiting multi-modal therapeutic options. Mitochondrial DNA (mtDNA) is an excellent resource for biomarker discovery because of its high copy number and increased mutational frequency in cancer cells. We examined if mtDNA mutations can be detected in circulating extracellular vesicles (EVs) of PDAC patients and used for discerning between cancer and non-cancer subjects. A greater yield of circulating EVs (~ 1.4 fold; p = 0.002) was obtained in PDAC patients (n = 20) than non-cancer (NC) individuals (n = 10). PDAC-EVs contained a higher quantity of total DNA (~ 5.5 folds; p = 0.0001) than NC-EVs and had greater enrichment of mtDNA (~ 14.02-fold; p = 0.0001). PDAC-EVs also had higher levels of cardiolipin (a mitochondrial inner-membrane phospholipid), suggestive of their mitochondrial origin. All mtDNA mutations in PDAC-EVs were unique and frequency was remarkably higher. Most mtDNA mutations (41.5%) in PDAC-EVs were in the respiratory complex-I (RCI) (ND1-ND6), followed by the RCIII gene (CYTB; 11.2%). Among the non-coding genes, D-Loop and RNR2 exhibited the most mutations (15.2% each). Altogether, our study establishes, for the first time, that mtDNA mutations can be detected in circulating EVs and potentially serve as a tool for reliable PDAC diagnosis.
Collapse
Affiliation(s)
- Kunwar Somesh Vikramdeo
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
| | - Shashi Anand
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
| | - Mohammad Aslam Khan
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
| | - Moh'd Khushman
- Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
- Division of Medical Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin J Heslin
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA
| | - Seema Singh
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, 36688, USA
| | - Ajay Pratap Singh
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA.
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA.
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, 36688, USA.
| | - Santanu Dasgupta
- Cancer Biology Program, Department of Pathology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, 1660 Springhill Avenue, Mobile, AL, 36604, USA.
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, 36617, USA.
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
24
|
Sui Z, Song X, Wu Y, Hou R, Liu J, Zhao B, Liang Z, Chen J, Zhang L, Zhang Y. The cytotoxicity of PM 2.5 and its effect on the secretome of normal human bronchial epithelial cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:75966-75977. [PMID: 35665889 DOI: 10.1007/s11356-022-20726-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
Exposure to airborne fine particulate matter (PM2.5) induced various adverse health effects, such as metabolic syndrome, systemic inflammation, and respiratory disease. Many works have studied the effects of PM2.5 exposure on cells through intracellular proteomics analyses. However, changes of the extracellular proteome under PM2.5 exposure and its correlation with PM2.5-induced cytotoxicity still remain unclear. Herein, the cytotoxicity of PM2.5 on normal human bronchial epithelia cells (BEAS-2B cells) was evaluated, and the secretome profile of BEAS-2B cells before and after PM2.5 exposure was investigated. A total of 83 proteins (58 upregulated and 25 downregulated) were differentially expressed in extracellular space after PM2.5 treatment. Notably, we found that PM2.5 promoted the release of several pro-apoptotic factors and induced dysregulated secretion of extracellular matrix (ECM) constituents, showing that the abnormal extracellular environment attributed to PM2.5-induced cell damage. This study provided a secretome data for the deep understanding of the molecular mechanism underlying PM2.5-caused human bronchial epithelia cell damage.
Collapse
Affiliation(s)
- Zhigang Sui
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiaoyao Song
- Environmental Assessment and Analysis Group, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yujie Wu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, 116024, China
| | - Rui Hou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| | - Jianhui Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| | - Zhen Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| | - Jiping Chen
- Environmental Assessment and Analysis Group, Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China.
| | - Yukui Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, 457 Zhongshan Road, Dalian, 116023, China
| |
Collapse
|
25
|
Zmigrodzka M, Witkowska-Pilaszewicz O, Pingwara R, Pawlak A, Winnicka A. Canine B Cell Lymphoma- and Leukemia-Derived Extracellular Vesicles Moderate Differentiation and Cytokine Production of T and B Cells In Vitro. Int J Mol Sci 2022; 23:ijms23179831. [PMID: 36077229 PMCID: PMC9456052 DOI: 10.3390/ijms23179831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/24/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are formed in physiological and pathological conditions by almost all mammalian cells. They are known as submicron “molecules” that transport and horizontally transfer their cargo from maternal cells to donor cells. Moreover, cancer cells produce tumor-derived EVs (TEVs), which are present in blood of patients with solid tumors and those with hematological malignancies. Their role in evading immune system surveillance and induction of immunosuppression in hematological cancer is limited. According to the authors’ best knowledge, there is no information about the impact of TEVs from canine lymphoma (CLBL-1) and leukemia (CLB70) on lymphocytes isolated from peripheral blood mononuclear cells (PBMCs). In conclusion, we demonstrate in in vitro experiments that CLBL-1 EVs and CLB70 EVs are effectively taken up by T and B lymphocytes. TEVs decrease the percentage of B lymphocytes and increase that of T lymphocytes, and change T cells’ phenotype into the effector memory (EM) or terminally differentiated effector memory (TEMRA) subtype after in vitro co-culturing. Moreover, CLBL70 EVs have pro-tumorogenic properties by inhibiting the production of CD8+IL-17+ cells.
Collapse
Affiliation(s)
- Magdalena Zmigrodzka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Olga Witkowska-Pilaszewicz
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Rafał Pingwara
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, CK Norwida 31, 50-375 Wroclaw, Poland
| | - Anna Winnicka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| |
Collapse
|
26
|
Lopez K, Camacho A, Jacquez Q, Amistadi MK, Medina S, Zychowski K. Lung-Based, Exosome Inhibition Mediates Systemic Impacts Following Particulate Matter Exposure. TOXICS 2022; 10:457. [PMID: 36006136 PMCID: PMC9413489 DOI: 10.3390/toxics10080457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Particulate matter (PM) exposure is a global health issue that impacts both urban and rural communities. Residential communities in the Southwestern United States have expressed concerns regarding the health impacts of fugitive PM from rural, legacy mine-sites. In addition, the recent literature suggests that exosomes may play a role in driving toxicological phenotypes following inhaled exposures. In this study, we assessed exosome-driven mechanisms and systemic health impacts following inhaled dust exposure, using a rodent model. Using an exosome inhibitor, GW4869 (10 μM), we inhibited exosome generation in the lungs of mice via oropharyngeal aspiration. We then exposed mice to previously characterized inhaled particulate matter (PM) from a legacy mine-site and subsequently assessed downstream behavioral, cellular, and molecular biomarkers in lung, serum, and brain tissue. Results indicated that CCL-2 was significantly upregulated in the lung tissue and downregulated in the brain (p < 0.05) following PM exposure. Additional experiments revealed cerebrovascular barrier integrity deficits and increased glial fibrillary acidic protein (GFAP) staining in the mine-PM exposure group, mechanistically dependent on exosome inhibition. An increased stress and anxiety response, based on the open-field test, was noted in the mine-PM exposure group, and subsequently mitigated with GW4869 intervention. Exosome lipidomics revealed 240 and eight significantly altered positive-ion lipids and negative-ion lipids, respectively, across the three treatment groups. Generally, phosphatidylethanolamine (PE) and phosphatidylcholine (PC) lipids were significantly downregulated in the PM group, compared to FA. In conclusion, these data suggest that systemic, toxic impacts of inhaled PM may be mechanistically dependent on lung-derived, circulating exosomes, thereby driving a systemic, proinflammatory phenotype.
Collapse
Affiliation(s)
- Keegan Lopez
- Department of Biology, College of Arts and Sciences, New Mexico Highlands University, Las Vegas, NM 88901, USA
| | - Alexandra Camacho
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| | - Quiteria Jacquez
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| | - Mary Kay Amistadi
- Arizona Laboratory for Emerging Contaminants, University of Arizona, Tucson, AZ 85721, USA
| | - Sebastian Medina
- Department of Biology, College of Arts and Sciences, New Mexico Highlands University, Las Vegas, NM 88901, USA
| | - Katherine Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
27
|
Uribe J, Traberg WC, Hama A, Druet V, Mohamed Z, Ooi A, Pappa AM, Huerta M, Inal S, Owens RM, Daniel S. Dual Mode Sensing of Binding and Blocking of Cancer Exosomes to Biomimetic Human Primary Stem Cell Surfaces. ACS Biomater Sci Eng 2021; 7:5585-5597. [PMID: 34802228 DOI: 10.1021/acsbiomaterials.1c01056] [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: 11/29/2022]
Abstract
Cancer-derived exosomes (cEXOs) facilitate transfer of information between tumor and human primary stromal cells, favoring cancer progression. Although the mechanisms used during this information exchange are still not completely understood, it is known that binding is the initial contact established between cEXOs and cells. Hence, studying binding and finding strategies to block it are of great therapeutic value. However, such studies are challenging for a variety of reasons, including the need for human primary cell culture, the difficulty in decoupling and isolating binding from internalization and cargo delivery, and the lack of techniques to detect these specific interactions. In this work, we created a supported biomimetic stem cell membrane incorporating membrane components from human primary adipose-derived stem cells (ADSCs). We formed the supported membrane on glass and on multielectrode arrays to offer the dual option of optical or electrical detection of cEXO binding to the membrane surface. Using our platform, we show that cEXOs bind to the stem cell membrane and that binding is blocked when an antibody to integrin β1, a component of ADSC surface, is exposed to the membrane surface prior to cEXOs. To test the biological outcome of blocking this interaction, we first confirm that adding cEXOs to cultured ADSCs leads to the upregulation of vascular endothelial growth factor, a measure of proangiogenic activity. Next, when ADSCs are first blocked with anti-integrin β1 and then exposed to cEXOs, the upregulation of proangiogenic activity and cell proliferation are significantly reduced. This biomimetic membrane platform is the first cell-free label-free in vitro platform for the recapitulation and study of cEXO binding to human primary stem cells with potential for therapeutic molecule screening as it is compatible with scale-up and multiplexing.
Collapse
Affiliation(s)
- Johana Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853-0001, United States
| | - Walther C Traberg
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Adel Hama
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 3955, Kingdom of Saudi Arabia
| | - Victor Druet
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 3955, Kingdom of Saudi Arabia
| | - Zeinab Mohamed
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853-0001, United States
| | - Amanda Ooi
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 3955, Kingdom of Saudi Arabia
| | - Anna-Maria Pappa
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom.,Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Miriam Huerta
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853-5201, United States
| | - Sahika Inal
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 3955, Kingdom of Saudi Arabia
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Susan Daniel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853-0001, United States.,School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853-5201, United States
| |
Collapse
|
28
|
|