51
|
Cvjetkovic A, Karimi N, Crescitelli R, Thorsell A, Taflin H, Lässer C, Lötvall J. Proteomic profiling of tumour tissue-derived extracellular vesicles in colon cancer. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e127. [PMID: 38939898 PMCID: PMC11080707 DOI: 10.1002/jex2.127] [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: 08/29/2022] [Revised: 07/19/2023] [Accepted: 10/23/2023] [Indexed: 06/29/2024]
Abstract
Colon cancer is one of the most commonly occurring tumours among both women and men, and over the past decades the incidence has been on the rise. As such, the need for biomarker identification as well as an understanding of the underlying disease mechanism has never been greater. Extracellular vesicles are integral mediators of cell-to-cell communication and offer a unique opportunity to study the machinery that drives disease progression, and they also function as vectors for potential biomarkers. Tumour tissue and healthy mucosal tissue from the colons of ten patients were used to isolate tissue-resident EVs that were subsequently subjected to global quantitative proteomic analysis through LC-MS/MS. In total, more than 2000 proteins were identified, with most of the common EV markers being among them. Bioinformatics revealed a clear underrepresentation of proteins involved in energy production and cellular adhesion in tumour EVs, while proteins involved in protein biosynthesis were overrepresented. Additionally, 53 membrane proteins were found to be significantly upregulated in tumour EVs. Among them were several proteins with enzymatic functions that degrade the extracellular matrix, and three of these, Fibroblast activating factor (FAP), Cell surface hyaluronidase (CEMIP2), as well as Ephrin receptor B3 (EPHB3), were validated and found to be consistent with the global quantitative results. These stark differences in the proteomes between healthy and cancerous tissue emphasise the importance of the interstitial vesicle secretome as a major player of disease development.
Collapse
Affiliation(s)
- Aleksander Cvjetkovic
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Nasibeh Karimi
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Rossella Crescitelli
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational Medicine, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University HospitalGothenburgSweden
| | - Annika Thorsell
- Proteomics Core Facility, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Helena Taflin
- Transplant Institute at Sahlgrenska University Hospital, Institute of Clinical SciencesSahlgrenska Academy at University of GothenburgGothenburgSweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
52
|
Wang X, Wang L, Lin H, Zhu Y, Huang D, Lai M, Xi X, Huang J, Zhang W, Zhong T. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front Oncol 2024; 14:1303335. [PMID: 38333685 PMCID: PMC10850354 DOI: 10.3389/fonc.2024.1303335] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vehicles (EVs) have received significant attention in recent times as emerging biomarkers and subjects of transformational studies. The three main branches of liquid biopsy have evolved from the three primary tumor liquid biopsy detection targets-CTC, ctDNA, and EVs-each with distinct benefits. CTCs are derived from circulating cancer cells from the original tumor or metastases and may display global features of the tumor. ctDNA has been extensively analyzed and has been used to aid in the diagnosis, treatment, and prognosis of neoplastic diseases. EVs contain tumor-derived material such as DNA, RNA, proteins, lipids, sugar structures, and metabolites. The three provide different detection contents but have strong complementarity to a certain extent. Even though they have already been employed in several clinical trials, the clinical utility of three biomarkers is still being studied, with promising initial findings. This review thoroughly overviews established and emerging technologies for the isolation, characterization, and content detection of CTC, ctDNA, and EVs. Also discussed were the most recent developments in the study of potential liquid biopsy biomarkers for cancer diagnosis, therapeutic monitoring, and prognosis prediction. These included CTC, ctDNA, and EVs. Finally, the potential and challenges of employing liquid biopsy based on CTC, ctDNA, and EVs for precision medicine were evaluated.
Collapse
Affiliation(s)
- Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Lijuan Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Haihong Lin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Yifan Zhu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mi Lai
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xuxiang Xi
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Wenjuan Zhang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
53
|
Maqsood Q, Sumrin A, Saleem Y, Wajid A, Mahnoor M. Exosomes in Cancer: Diagnostic and Therapeutic Applications. Clin Med Insights Oncol 2024; 18:11795549231215966. [PMID: 38249520 PMCID: PMC10799603 DOI: 10.1177/11795549231215966] [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: 06/22/2023] [Accepted: 10/29/2023] [Indexed: 01/23/2024] Open
Abstract
Small extracellular vesicles called exosomes are produced by cells and contain a range of biomolecules, including proteins, lipids, and nucleic acids. Exosomes have been implicated in the development and spread of cancer, and recent studies have shown that their contents may be exploited as biomarkers for early detection and ongoing surveillance of the disease. In this review article, we summarize the current knowledge on exosomes as biomarkers of cancer. We discuss the various methods used for exosome isolation and characterization, as well as the different types of biomolecules found within exosomes that are relevant for cancer diagnosis and prognosis. We also highlight recent studies that have demonstrated the utility of exosomal biomarkers in different types of cancer, such as lung cancer, breast cancer, and pancreatic cancer. Overall, exosomes show great promise as noninvasive biomarkers for cancer detection and monitoring. Exosomes have the ability to transform cancer diagnostic and therapeutic paradigms, providing promise for more efficient and individualized. This review seeks to serve as an inspiration for new ideas and research in the never-ending fight against cancer. Moreover, further studies are needed to validate their clinical utility and establish standardized protocols for their isolation and analysis. With continued research and development, exosomal biomarkers have the potential to revolutionize cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Quratulain Maqsood
- Department of Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Aleena Sumrin
- Department of Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Yasar Saleem
- Department of Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Laboratories Complex Lahore, Lahore, Pakistan
| | - Abdul Wajid
- Department of Biotechnology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, Pakistan
| | - Muhammada Mahnoor
- Department of Rehabilitation Science, The University of Lahore, Lahore, Pakistan
| |
Collapse
|
54
|
Sun Z, Zhang B, Tu H, Pan C, Chai Y, Chen W. Advances in colorimetric biosensors of exosomes: novel approaches based on natural enzymes and nanozymes. NANOSCALE 2024; 16:1005-1024. [PMID: 38117141 DOI: 10.1039/d3nr05459d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Exosomes are 30-150 nm vesicles derived from diverse cell types, serving as one of the most important biomarkers for early diagnosis and prognosis of diseases. However, the conventional detection method for exosomes faces significant challenges, such as unsatisfactory sensitivity, complicated operation, and the requirement of complicated devices. In recent years, colorimetric exosome biosensors with a visual readout underwent rapid development due to the advances in natural enzyme-based assays and the integration of various types of nanozymes. These synthetic nanomaterials show unique physiochemical properties and catalytic abilities, enabling the construction of exosome colorimetric biosensors with novel principles. This review will illustrate the reaction mechanisms and properties of natural enzymes and nanozymes, followed by a detailed introduction of the recent advances in both types of enzyme-based colorimetric biosensors. A comparison between natural enzymes and nanozymes is made to provide insights into the research that improves the sensitivity and convenience of assays. Finally, the advantages, challenges, and future directions of enzymes as well as exosome colorimetric biosensors are highlighted, aiming at improving the overall performance from different approaches.
Collapse
Affiliation(s)
- Zhonghao Sun
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Hangjia Tu
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Chuye Pan
- College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Wenwen Chen
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
55
|
Fais S, Logozzi M. The Diagnostic and Prognostic Value of Plasmatic Exosome Count in Cancer Patients and in Patients with Other Pathologies. Int J Mol Sci 2024; 25:1049. [PMID: 38256122 PMCID: PMC10816819 DOI: 10.3390/ijms25021049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The extent of both scientific articles and reviews on extracellular vesicles (EVs) has grown impressively over the last few decades [...].
Collapse
Affiliation(s)
- Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
- ExoLab Italia, Tecnopolo d’Abruzzo, 67100 L’Aquila, Italy
| | - Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
56
|
Hánělová K, Raudenská M, Masařík M, Balvan J. Protein cargo in extracellular vesicles as the key mediator in the progression of cancer. Cell Commun Signal 2024; 22:25. [PMID: 38200509 PMCID: PMC10777590 DOI: 10.1186/s12964-023-01408-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/24/2023] [Indexed: 01/12/2024] Open
Abstract
Exosomes are small vesicles of endosomal origin that are released by almost all cell types, even those that are pathologically altered. Exosomes widely participate in cell-to-cell communication via transferring cargo, including nucleic acids, proteins, and other metabolites, into recipient cells. Tumour-derived exosomes (TDEs) participate in many important molecular pathways and affect various hallmarks of cancer, including fibroblasts activation, modification of the tumour microenvironment (TME), modulation of immune responses, angiogenesis promotion, setting the pre-metastatic niche, enhancing metastatic potential, and affecting therapy sensitivity and resistance. The unique exosome biogenesis, composition, nontoxicity, and ability to target specific tumour cells bring up their use as promising drug carriers and cancer biomarkers. In this review, we focus on the role of exosomes, with an emphasis on their protein cargo, in the key mechanisms promoting cancer progression. We also briefly summarise the mechanism of exosome biogenesis, its structure, protein composition, and potential as a signalling hub in both normal and pathological conditions. Video Abstract.
Collapse
Affiliation(s)
- Klára Hánělová
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Martina Raudenská
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
| | - Michal Masařík
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, Vestec, CZ-252 50, Czech Republic
| | - Jan Balvan
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, CZ-625 00, Czech Republic.
| |
Collapse
|
57
|
Wang Y, Lu C, Guo S, Guo Y, Wei T, Chen Q. Leafhopper salivary vitellogenin mediates virus transmission to plant phloem. Nat Commun 2024; 15:3. [PMID: 38167823 PMCID: PMC10762104 DOI: 10.1038/s41467-023-43488-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024] Open
Abstract
Salivary effectors of piercing-sucking insects can suppress plant defense to promote insect feeding, but it remains largely elusive how they facilitate plant virus transmission. Leafhopper Nephotettix cincticeps transmits important rice reovirus via virus-packaging exosomes released from salivary glands and then entering the rice phloem. Here, we report that intact salivary vitellogenin of N. cincticeps (NcVg) is associated with the GTPase Rab5 of N. cincticeps (NcRab5) for release from salivary glands. In virus-infected salivary glands, NcVg is upregulated and packaged into exosomes mediated by virus-induced NcRab5, subsequently entering the rice phloem. The released NcVg inherently suppresses H2O2 burst of rice plants by interacting with rice glutathione S-transferase F12, an enzyme catalyzing glutathione-dependent oxidation, thus facilitating leafhoppers feeding. When leafhoppers transmit virus, virus-upregulated NcVg thus promotes leafhoppers feeding and enhances viral transmission. Taken together, the findings provide evidence that viruses exploit insect exosomes to deliver virus-hijacked effectors for efficient transmission.
Collapse
Affiliation(s)
- Yanfei Wang
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Chengcong Lu
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Shude Guo
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yuxin Guo
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Taiyun Wei
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Qian Chen
- Vector-borne Virus Research Center, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China.
| |
Collapse
|
58
|
Mojtaba Mousavi S, Alireza Hashemi S, Yari Kalashgrani M, Rahmanian V, Riazi M, Omidifar N, Hamed Althomali R, Rahman MM, Chiang WH, Gholami A. Recent Progress in Prompt Molecular Detection of Exosomes Using CRISPR/Cas and Microfluidic-Assisted Approaches Toward Smart Cancer Diagnosis and Analysis. ChemMedChem 2024; 19:e202300359. [PMID: 37916531 DOI: 10.1002/cmdc.202300359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/03/2023]
Abstract
Exosomes are essential indicators of molecular mechanisms involved in interacting with cancer cells and the tumor environment. As nanostructures based on lipids and nucleic acids, exosomes provide a communication pathway for information transfer by transporting biomolecules from the target cell to other cells. Importantly, these extracellular vesicles are released into the bloodstream by the most invasive cells, i. e., cancer cells; in this way, they could be considered a promising specific biomarker for cancer diagnosis. In this matter, CRISPR-Cas systems and microfluidic approaches could be considered practical tools for cancer diagnosis and understanding cancer biology. CRISPR-Cas systems, as a genome editing approach, provide a way to inactivate or even remove a target gene from the cell without affecting intracellular mechanisms. These practical systems provide vital information about the factors involved in cancer development that could lead to more effective cancer treatment. Meanwhile, microfluidic approaches can also significantly benefit cancer research due to their proper sensitivity, high throughput, low material consumption, low cost, and advanced spatial and temporal control. Thereby, employing CRISPR-Cas- and microfluidics-based approaches toward exosome monitoring could be considered a valuable source of information for cancer therapy and diagnosis. This review assesses the recent progress in these promising diagnosis approaches toward accurate cancer therapy and in-depth study of cancer cell behavior.
Collapse
Affiliation(s)
- Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City, 106335, Taiwan
| | - Seyyed Alireza Hashemi
- Health Policy Research Center, Health Institute, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Vahid Rahmanian
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, Lodz, 90-363, Poland
| | - Mohsen Riazi
- Biotechnology Research Center, Shiraz University of Medical Science, Shiraz, 71468-64685, Iran
| | - Navid Omidifar
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammed M Rahman
- Center of Excellence for Advanced Materials Research (CEAMR) & Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City, 106335, Taiwan
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Science, Shiraz, 71468-64685, Iran
| |
Collapse
|
59
|
Rodríguez-Zorrilla S, Lorenzo-Pouso AI, Fais S, Logozzi MA, Mizzoni D, Di Raimo R, Giuliani A, García-García A, Pérez-Jardón A, Ortega KL, Martínez-González Á, Pérez-Sayáns M. Increased Plasmatic Levels of Exosomes Are Significantly Related to Relapse Rate in Patients with Oral Squamous Cell Carcinoma: A Cohort Study. Cancers (Basel) 2023; 15:5693. [PMID: 38067397 PMCID: PMC10705147 DOI: 10.3390/cancers15235693] [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: 09/24/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is characterized by an immunosuppressive tumor microenvironment. Their plasma-derived exosomes deliver immunomodulatory molecules and cargo that correlate significantly with clinical parameters. This study aims to assess the exosomal profile as a potential tool for early detection of relapse and long-term outcomes in OSCC patients undergoing conventional therapy. METHODS 27 OSCC patients with a median 38-month follow-up were included in this study. The relationship between NTA-derived parameters and clinical pathological parameters was examined, and receiver operating characteristic (ROC) curves were utilized to evaluate the diagnostic efficacy of these values in detecting cancer relapse. RESULTS Plasmatic levels of exosomes prior to surgery showed a drastic reduction after surgical intervention (8.08E vs. 1.41 × 109 particles/mL, p = 0.006). Postsurgical concentrations of exosomes were higher in patients who experienced relapse compared to those who remained disease-free (2.97 × 109 vs. 1.11 × 109 particles/mL, p = 0.046). Additionally, patients who relapsed exhibited larger exosome sizes after surgery (141.47 vs. 132.31 nm, p = 0.03). Patients with lower concentrations of exosomes prior to surgery demonstrated better disease-free survival compared to those with higher levels (p = 0.012). ROC analysis revealed an area under the curve of 0.82 for presurgical exosome concentration in identifying relapse. CONCLUSIONS Presurgical exosomal plasmatic levels serve as independent predictors of early recurrence and survival in OSCC. All in all, our findings indicate that the detection of peripheral exosomes represents a novel tool for the clinical management of OSCC, with potential implications for prognosis assessment.
Collapse
Affiliation(s)
- Samuel Rodríguez-Zorrilla
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
| | - Alejandro I. Lorenzo-Pouso
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
- ORALRES Group, Health Research Institute of Santiago de Compostela (FIDIS), 15782 Santiago de Compostela, Spain
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (S.F.); (M.A.L.)
| | - Maria A. Logozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (S.F.); (M.A.L.)
| | - Davide Mizzoni
- ExoLab Italia, Tecnopolo d’Abruzzo, 67100 L’Aquila, Italy; (D.M.); (R.D.R.)
| | - Rossella Di Raimo
- ExoLab Italia, Tecnopolo d’Abruzzo, 67100 L’Aquila, Italy; (D.M.); (R.D.R.)
| | - Alessandro Giuliani
- Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Abel García-García
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
- ORALRES Group, Health Research Institute of Santiago de Compostela (FIDIS), 15782 Santiago de Compostela, Spain
| | - Alba Pérez-Jardón
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
- ORALRES Group, Health Research Institute of Santiago de Compostela (FIDIS), 15782 Santiago de Compostela, Spain
| | - Karem L. Ortega
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
- School of Dentistry, Department of Oral Pathology, University of São Paulo, Av. Lineu Prestes, 2227, Cidade Universitária São Paulo, Sao Paulo 05508-000, Brazil
| | - Ángel Martínez-González
- Endocrinology and Nutrition Service, Complejo Hospitalario Universitario de Pontevedra, Mourente S/N, 36472 Pontevedra, Spain;
| | - Mario Pérez-Sayáns
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.R.-Z.); (A.G.-G.); (A.P.-J.); (K.L.O.); (M.P.-S.)
- ORALRES Group, Health Research Institute of Santiago de Compostela (FIDIS), 15782 Santiago de Compostela, Spain
- Institute of Materials (IMATUS), Avenida do Mestre Mateo, 25, 15782 Santiago de Compostela, Spain
| |
Collapse
|
60
|
Matejova J, Fecskeova LK, Slovinska L, Harvanova D, Spakova T, Bzdilova J. Plasma-derived extracellular vesicle surface markers CD45, CD326 and CD56 correlate with the stage of osteoarthritis: a primary study of a novel and promising diagnostic tool of the disease. Sci Rep 2023; 13:20071. [PMID: 37973964 PMCID: PMC10654566 DOI: 10.1038/s41598-023-47074-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: 07/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Recently, there is a growing interest in the research based on extracellular vesicles (EVs) which represent paracrine factors secreted by almost all cell types. Both, normal and pathological cells are able to release various types of EVs with different physiological properties, functions and compositions. EVs play an important role in intercellular communication, mechanism and tissue repair. Moreover, EVs could help not only in the treatment of diseases but also in their diagnostics. This work focused on the evaluation of the potential of EVs being used as biomarkers for the diagnosis of osteoarthritis (OA) based on a comparison of the composition of EVs separated from platelet-poor plasma (PPP) of healthy donors and OA patients at different stages of OA. OA is established as a complex syndrome with extensive impact on multiple tissues within the synovial joint. It is a chronic disease of musculoskeletal system that mainly affects the elderly. Depending on the use of the Kellgren-Lawrence classification system, there are four grades of OA which have a negative impact on patients' quality of life. It is very difficult to detect OA in its early stages, so it is necessary to find a new diagnostic method for its timely detection. PPP samples were prepared from whole blood. PPP-EVs were separated from 3 groups of donors-healthy control, early stage OA, end-stage OA, and their content was compared and correlated. EVs from PPP were separated by size exclusion chromatography and characterized in terms of their size, yield and purity by NTA, western blotting, ELISA and flow cytometry. Detection of surface markers expression in EVs was performed using MACSPlex approach. Inflammatory and growth factors in EVs were analysed using MAGPix technology. Our study confirmed significant differences between EVs surface markers of patients and healthy controls correlating with the age of donor (CD63, CD31 and ROR1) and stage of OA (CD45, CD326 and CD56), respectively. Circulating EVs have been under extensive investigation for their capability to predict OA pathology diagnosis as potential targets for biomarker discovery. Taken together, obtained results indicated that PPP-EVs surface markers could be used as potential biomarkers in the early diagnosis of OA.
Collapse
Affiliation(s)
- Jana Matejova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Livia K Fecskeova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Lucia Slovinska
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Denisa Harvanova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Timea Spakova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia
| | - Jana Bzdilova
- Associated Tissue Bank, Faculty of Medicine, P. J. Safarik University and L. Pasteur University Hospital in Kosice, Tr. SNP 1, 04011, Kosice, Slovakia.
| |
Collapse
|
61
|
Altıntaş Ö, Saylan Y. Exploring the Versatility of Exosomes: A Review on Isolation, Characterization, Detection Methods, and Diverse Applications. Anal Chem 2023; 95:16029-16048. [PMID: 37874907 DOI: 10.1021/acs.analchem.3c02224] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Extracellular vesicles (EVs) are crucial mediators of intercellular communication and can be classified based on their physical properties, biomolecular structure, and origin. Among EVs, exosomes have garnered significant attention due to their potential as therapeutic and diagnostic tools. Exosomes are released via fusion of multivesicular bodies on plasma membranes and can be isolated from various biofluids using methods such as differential ultracentrifugation, immune affinity capture, ultrafiltration, and size exclusion chromatography. Herein, an overview of different techniques for exosome characterization and isolation, as well as the diverse applications of exosome detection, including their potential use in drug delivery and disease diagnosis, is provided. Additionally, we discuss the emerging field of exosome detection by sensors, which offers an up-and-coming avenue for point-of-care diagnostic tools development. Overall, this review aims to provide a exhaustive and up-to-date summary of the current state of exosome research.
Collapse
Affiliation(s)
- Özge Altıntaş
- Hacettepe University, Department of Chemistry, 06800 Ankara, Turkey
| | - Yeşeren Saylan
- Hacettepe University, Department of Chemistry, 06800 Ankara, Turkey
| |
Collapse
|
62
|
Dey D, Ghosh S, Mirgh D, Panda SP, Jha NK, Jha SK. Role of exosomes in prostate cancer and male fertility. Drug Discov Today 2023; 28:103791. [PMID: 37777169 DOI: 10.1016/j.drudis.2023.103791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Prostate cancer (PCa) is the second most common and fifth most aggressive neoplasm among men worldwide. In the last decade, extracellular vesicle (EV) research has decoded multiple unsolved cancer-related mysteries. EVs can be classified as microvesicles, apoptotic bodies, and exosomes, among others. Exosomes play a key role in cellular signaling. Their internal cargos (nucleic acids, proteins, lipids) influence the recipient cell. In PCa, the exosome is the regulator of cancer progression. It is also a promising theranostics tool for PCa. Moreover, exosomes have strong participation in male fertility complications. This review aims to highlight the exosome theranostics signature in PCa and its association with male fertility.
Collapse
Affiliation(s)
- Dwaipayan Dey
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Rahara, West Bengal 700118, India
| | - Srestha Ghosh
- Department of Microbiology, Lady Brabourne College, Kolkata 700017, West Bengal, India
| | - Divya Mirgh
- Johns Hopkins University, Baltimore, MD 21218, USA
| | - Siva Parsad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal, University, Dehradun, India.
| |
Collapse
|
63
|
Bano A, Vats R, Verma D, Yadav P, Kamboj M, Bhardwaj R. Exploring salivary exosomes as early predictors of oral cancer in susceptible tobacco consumers: noninvasive diagnostic and prognostic applications. J Cancer Res Clin Oncol 2023; 149:15781-15793. [PMID: 37668794 DOI: 10.1007/s00432-023-05343-4] [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: 06/20/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Salivary exosome analysis provides a noninvasive and comprehensive approach with potential applications in oral cancer diagnosis and prognosis. The early detection of oral cancer has remained a critical concern in enhancing the quality of life, especially for individuals who consume tobacco and are at greater risk of developing the disease. The current study investigates the potential of salivary exosomes in screening smokers for early signs and transformations of oral cancer. METHODS Morphological characterization of salivary exosomes among three study groups (non-smokers as control, smokers as high-risk tobacco consumers, and Oral cancer) (n = 120) was carried out through dynamic light scattering, and nanoparticle tracking analysis. For molecular characterization, EXOCET and Fourier transform infrared spectroscopy methods were utilized. The expression of the exosomal surface protein CD63 was evaluated using Western blotting. RESULTS Salivary exosomes exhibit noticeable differences in size between control group and tobacco consumers. The differentiation extended beyond exosome size and included variations in concentration and bio-molecular composition, as determined by FTIR screening. Tobacco consumers and oral cancer groups showed significantly larger and more concentrated exosomes compared to the healthy group. CONCLUSION Our study provides strong evidence that the properties of salivary exosomes can serve as reliable noninvasive biomarkers for distinguishing tobacco consumers from non-smokers and oral cancer patients. Our results underscore the potential of exosome-based diagnostics in early oral cancer detection for high-risk individuals. The larger size and higher concentration of exosomes in tobacco consumers indicate early changes in cell secretions associated with the transformation from healthy to abnormal cells.
Collapse
Affiliation(s)
- Afsareen Bano
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 122001, India
| | - Ravina Vats
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 122001, India
| | - Deepika Verma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Pooja Yadav
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 122001, India
| | - Mala Kamboj
- Department of Oral Pathology, Postgraduate Institute of Dental Sciences, Rohtak, Haryana, 124001, India
| | - Rashmi Bhardwaj
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 122001, India.
| |
Collapse
|
64
|
Hadvina R, Lotfy Khaled M, Akoto T, Zhi W, Karamichos D, Liu Y. Exosomes and their miRNA/protein profile in keratoconus-derived corneal stromal cells. Exp Eye Res 2023; 236:109642. [PMID: 37714423 PMCID: PMC10842962 DOI: 10.1016/j.exer.2023.109642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
Keratoconus (KC) is a corneal thinning disorder and a leading cause of corneal transplantation worldwide. Exosomes are small, secreted extracellular vesicles (30-150 nm) that mediate cellular communication via their protein, lipid, and nucleic acid content. We aimed to characterize the exosomes secreted by primary corneal fibroblasts from subjects with or without KC. Using human keratoconus stromal fibroblast cells (HKC, n = 4) and healthy stromal fibroblasts (HCF, n = 4), we collected and isolated exosomes using serial ultracentrifugation. Using nanoparticle tracking analysis (NTA) with ZetaView®, we compared the size and concentration of isolated exosomes. Different exosomal markers were identified and quantified using a transmission electron microscope (TEM) (CD81) and Western blot (CD9 and CD63). Exosomal miRNA profiles were determined by qRT-PCR using Exiqon Human panel I miRNA assays of 368 pre-selected miRNAs. Proteomic profiles were determined using a label-free spectral counting method with mass spectrometry. Differential expression analysis for miRNAs and proteins was done using student's t-test with a significance cutoff of p-value ≤0.05. We successfully characterized exosomes isolated from HCFs using several complementary techniques. We found no significant differences in the size, quantity, or morphology between exosomes secreted by HCFs with or without KC. Expression of CD81 was confirmed by immuno-EM, and expression of CD63 and CD9 with western blots in all exosome samples. We detected the expression of 72-144 miRNAs (threshold cycle Ct < 36) in all exosome samples. In HKC-derived exosome samples, miR-328-3p, miR-532-5p, miR-345-5p, and miR-424-5p showed unique expression, while let-7c-5p and miR-665 have increased expression. Protein profiling identified 157 proteins in at least half of the exosome samples, with 38 known exosomal proteins. We identified 12 up- and 2 down-regulated proteins in HKC-derived exosomes. The proteins are involved in membrane-bounded vesicles, cytoskeletal, calcium binding, and nucleotide binding. These proteins are predicted to be regulated by NRF2, miR-205, and TGF-β1, which are involved in KC pathogenesis. We successfully characterized the HKC-derived exosomes and profiled their miRNA and protein contents, suggesting their potential role in KC development. Further studies are necessary to determine if and how these exosomes with differential protein/miRNA profiles contribute to the pathogenesis of KC.
Collapse
Affiliation(s)
- Rachel Hadvina
- Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, 30912, USA; Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Mariam Lotfy Khaled
- Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, 30912, USA; Department of Biochemistry, Cairo University, Egypt
| | - Theresa Akoto
- Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, 30912, USA
| | - Wenbo Zhi
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.
| | - Yutao Liu
- Department of Cellular Biology & Anatomy, Augusta University, Augusta, GA, 30912, USA; Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; James & Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
65
|
Nejatie A, Yee SS, Jeter A, Saragovi HU. The cancer glycocode as a family of diagnostic biomarkers, exemplified by tumor-associated gangliosides. Front Oncol 2023; 13:1261090. [PMID: 37954075 PMCID: PMC10637394 DOI: 10.3389/fonc.2023.1261090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023] Open
Abstract
One unexploited family of cancer biomarkers comprise glycoproteins, carbohydrates, and glycolipids (the Tumor Glycocode).A class of glycolipid cancer biomarkers, the tumor-marker gangliosides (TMGs) are presented here as potential diagnostics for detecting cancer, especially at early stages, as the biological function of TMGs makes them etiological. We propose that a quantitative matrix of the Cancer Biomarker Glycocode and artificial intelligence-driven algorithms will expand the menu of validated cancer biomarkers as a step to resolve some of the challenges in cancer diagnosis, and yield a combination that can identify a specific cancer, in a tissue-agnostic manner especially at early stages, to enable early intervention. Diagnosis is critical to reducing cancer mortality but many cancers lack efficient and effective diagnostic tests, especially for early stage disease. Ideal diagnostic biomarkers are etiological, samples are preferably obtained via non-invasive methods (e.g. liquid biopsy of blood or urine), and are quantitated using assays that yield high diagnostic sensitivity and specificity for efficient diagnosis, prognosis, or predicting response to therapy. Validated biomarkers with these features are rare. While the advent of proteomics and genomics has led to the identification of a multitude of proteins and nucleic acid sequences as cancer biomarkers, relatively few have been approved for clinical use. The use of multiplex arrays and artificial intelligence-driven algorithms offer the option of combining data of known biomarkers; however, for most, the sensitivity and the specificity are below acceptable criteria, and clinical validation has proven difficult. One strategic solution to this problem is to expand the biomarker families beyond those currently exploited. One unexploited family of cancer biomarkers comprise glycoproteins, carbohydrates, and glycolipids (the Tumor Glycocode). Here, we focus on a family of glycolipid cancer biomarkers, the tumor-marker gangliosides (TMGs). We discuss the diagnostic potential of TMGs for detecting cancer, especially at early stages. We include prior studies from the literature to summarize findings for ganglioside quantification, expression, detection, and biological function and its role in various cancers. We highlight the examples of TMGs exhibiting ideal properties of cancer diagnostic biomarkers, and the application of GD2 and GD3 for diagnosis of early stage cancers with high sensitivity and specificity. We propose that a quantitative matrix of the Cancer Biomarker Glycocode and artificial intelligence-driven algorithms will expand the menu of validated cancer biomarkers as a step to resolve some of the challenges in cancer diagnosis, and yield a combination that can identify a specific cancer, in a tissue-agnostic manner especially at early stages, to enable early intervention.
Collapse
Affiliation(s)
- Ali Nejatie
- Center for Translational Research, Lady Davis Research Institute-Jewish General Hospital, Montreal, QC, Canada
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Samantha S. Yee
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, United States
| | | | - Horacio Uri Saragovi
- Center for Translational Research, Lady Davis Research Institute-Jewish General Hospital, Montreal, QC, Canada
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Ophthalmology and Vision Science, McGill University, Montreal, QC, Canada
| |
Collapse
|
66
|
Kumar S, Dhar R, Kumar LBSS, Shivji GG, Jayaraj R, Devi A. Theranostic signature of tumor-derived exosomes in cancer. Med Oncol 2023; 40:321. [PMID: 37798480 DOI: 10.1007/s12032-023-02176-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
Cancer is the most challenging global health crisis. In the recent times, studies on extracellular vesicles (EVs) are adding a new chapter to cancer research and reports on EVs explores cancer in a new dimension. Exosomes are a group of subpopulations of EVs. It originates from the endosomes and carries biologically active molecules to the neighboring cells which in turn transforms the recipient cell activity. In general, it plays a role in cellular communication. The correlation between exosomes and cancer is fascinating. Tumor-derived exosomes (TEXs) play a dynamic role in cancer progression and are associated with uncontrolled cell growth, angiogenesis, immune suppression, and metastasis. Its molecular cargo is an excellent source of cancer biomarkers. Several advanced molecular profiling approaches assist in exploring the TEXs in depth. This paves the way for a strong foundation for identifying and detecting more specific and efficient biomarkers. TEXs are also gaining importance in scientific society for its role in cancer therapy and several clinical trials based on TEXs is a proof of its significance. In this review, we have highlighted the role of TEXs in mediating immune cell reprogramming, cancer development, metastasis, EMT, organ-specific metastasis, and its clinical significance in cancer theranostics. TEXs profiling is an effective method to understand the complications associated with cancer leading to good health and well-being of the individual and society as a whole.
Collapse
Affiliation(s)
- Samruti Kumar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Lokesh Babu Sirkali Suresh Kumar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Gauresh Gurudas Shivji
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, 28, Sonipat, 131001, India
- Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT, 0909, Australia
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
67
|
Campos A, Burgos-Ravanal R, Lobos-González L, Huilcamán R, González MF, Díaz J, Verschae AC, Acevedo JP, Carrasco M, Sepúlveda F, Jeldes E, Varas-Godoy M, Leyton L, Quest AF. Caveolin-1-dependent tenascin C inclusion in extracellular vesicles is required to promote breast cancer cell malignancy. Nanomedicine (Lond) 2023; 18:1651-1668. [PMID: 37929694 DOI: 10.2217/nnm-2023-0143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
Background: Elevated expression of CAV1 in breast cancer increases tumor progression. Extracellular vesicles (EVs) from CAV1-expressing MDA-MB-231 breast cancer cells contain Tenascin C (TNC), but the relevance of TNC remained to be defined. Methods: EVs were characterized by nanotracking analysis, microscopy and western blotting. The uptake of EVs by cells was studied using flow cytometry. The effects of EVs on breast cancer cells were tested in migration, invasion, colony formation and in vivo assays. Results: EVs were taken up by cells; however, only those containing TNC promoted invasiveness. In vivo, EVs lacking TNC ceased to promote tumor growth. Conclusion: CAV1 and TNC contained in breast cancer cell-derived EVs were identified as proteins that favor progression of breast cancer.
Collapse
Affiliation(s)
- America Campos
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, 8340148, Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, Scotland
| | - Renato Burgos-Ravanal
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| | - Lorena Lobos-González
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, 7610615, Chile
| | - Ricardo Huilcamán
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| | - María Fernanda González
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| | - Jorge Díaz
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| | - Albano Cáceres Verschae
- Laboratorio de Biología Celular del Cáncer, CEBICEM, Universidad San Sebastián, Santiago, 7510157, Chile
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Juan Pablo Acevedo
- Center of Interventional Medicine for Precision & Advanced Cellular Therapy (IMPACT), Santiago, 8331150, Chile
| | - Macarena Carrasco
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, 8340148, Chile
| | - Francisca Sepúlveda
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, 8340148, Chile
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, 7610615, Chile
| | - Emanuel Jeldes
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, 8340148, Chile
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, Scotland
| | - Manuel Varas-Godoy
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago, 8340148, Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
- Laboratorio de Biología Celular del Cáncer, CEBICEM, Universidad San Sebastián, Santiago, 7510157, Chile
| | - Lisette Leyton
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| | - Andrew Fg Quest
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, 8380492, Universidad de Chile
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago, 8380492, Chile
| |
Collapse
|
68
|
Liguori GL, Kralj-Iglič V. Pathological and Therapeutic Significance of Tumor-Derived Extracellular Vesicles in Cancer Cell Migration and Metastasis. Cancers (Basel) 2023; 15:4425. [PMID: 37760395 PMCID: PMC10648223 DOI: 10.3390/cancers15184425] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
The infiltration of primary tumors and metastasis formation at distant sites strongly impact the prognosis and the quality of life of cancer patients. Current therapies including surgery, radiotherapy, and chemotherapy are limited in targeting the complex cell migration mechanisms responsible for cancer cell invasiveness and metastasis. A better understanding of these mechanisms and the development of new therapies are urgently needed. Extracellular vesicles (EVs) are lipid-enveloped particles involved in inter-tissue and inter-cell communication. This review article focuses on the impact of EVs released by tumor cells, specifically on cancer cell migration and metastasis. We first introduce cell migration processes and EV subtypes, and we give an overview of how tumor-derived EVs (TDEVs) may impact cancer cell migration. Then, we discuss ongoing EV-based cancer therapeutic approaches, including the inhibition of general EV-related mechanisms as well as the use of EVs for anti-cancer drug delivery, focusing on the harnessing of TDEVs. We propose a protein-EV shuttle as a route alternative to secretion or cell membrane binding, influencing downstream signaling and the final effect on target cells, with strong implications in tumorigenesis. Finally, we highlight the pitfalls and limitations of therapeutic EV exploitation that must be overcome to realize the promise of EVs for cancer therapy.
Collapse
Affiliation(s)
- Giovanna L. Liguori
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati-Traverso”, National Research Council (CNR) of Italy, 80131 Naples, Italy
| | - Veronika Kralj-Iglič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
69
|
Garcia NA, Mellergaard M, Gonzalez-King H, Salomon C, Handberg A. Comprehensive Strategy for Identifying Extracellular Vesicle Surface Proteins as Biomarkers for Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:13326. [PMID: 37686134 PMCID: PMC10487973 DOI: 10.3390/ijms241713326] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver disorder that has become a global health concern due to its increasing prevalence. There is a need for reliable biomarkers to aid in the diagnosis and prognosis of NAFLD. Extracellular vesicles (EVs) are promising candidates in biomarker discovery, as they carry proteins that reflect the pathophysiological state of the liver. In this review, we developed a list of EV proteins that could be used as diagnostic biomarkers for NAFLD. We employed a multi-step strategy that involved reviewing and comparing various sources of information. Firstly, we reviewed papers that have studied EVs proteins as biomarkers in NAFLD and papers that have studied circulating proteins as biomarkers in NAFLD. To further identify potential candidates, we utilized the EV database Vesiclepedia.org to qualify each protein. Finally, we consulted the Human Protein Atlas to search for candidates' localization, focusing on membrane proteins. By integrating these sources of information, we developed a comprehensive list of potential EVs membrane protein biomarkers that could aid in diagnosing and monitoring NAFLD. In conclusion, our multi-step strategy for identifying EV-based protein biomarkers for NAFLD provides a comprehensive approach that can also be applied to other diseases. The protein candidates identified through this approach could have significant implications for the development of non-invasive diagnostic tests for NAFLD and improve the management and treatment of this prevalent liver disorder.
Collapse
Affiliation(s)
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg Hobrovej 18-22, 9000 Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Hernan Gonzalez-King
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD 4029, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg Hobrovej 18-22, 9000 Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, 9000 Aalborg, Denmark
| |
Collapse
|
70
|
Sareen N, Srivastava A, Alagarsamy KN, Lionetti V, Dhingra S. Stem cells derived exosomes and biomaterials to modulate autophagy and mend broken hearts. Biochim Biophys Acta Mol Basis Dis 2023:166806. [PMID: 37437748 DOI: 10.1016/j.bbadis.2023.166806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023]
Abstract
Autophagy maintains cellular homeostasis and plays a crucial role in managing pathological conditions including ischemic myocardial injury leading to heart failure (HF). Despite treatments, no intervention can replace lost cardiomyocytes. Stem cell therapy offers potential for post-myocardial infarction repair but struggles with poor cell retention due to immune rejection. In the search for effective therapies, stem cell-derived extracellular vesicles (EVs), especially exosomes, have emerged as promising tools. These tiny bioactive molecule carriers play vital roles in intercellular communication and tissue engineering. They offer numerous therapeutic benefits including modulating immune responses, promoting tissue repair, and boosting angiogenesis. Additionally, biomaterials provide a conducive 3D microenvironment for cell, exosome, and biomolecule delivery, and enhance heart muscle strength, making it a comprehensive cardiac repair strategy. In this regard, the current review delves into the intricate application of extracellular vesicles (EVs) and biomaterials for managing autophagy in the heart muscle during cardiac injury. Central to our investigation is the exploration of how these elements interact within the context of cardiac repair and regeneration. Additionally, this review also casts light on the formidable challenges that plague this field, such as the issues of safety, efficacy, controlled delivery, and acceptance of these therapeutic strategies for effective clinical translation. Addressing these challenges is crucial for unlocking the full therapeutic potential of EV and biomaterial-based therapies and ensuring their successful translation from bench to bedside.
Collapse
Affiliation(s)
- Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada; Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Abhay Srivastava
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Keshav Narayan Alagarsamy
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada.
| |
Collapse
|
71
|
Jiang X, Wu S, Hu C. A narrative review of the role of exosomes and caveolin-1 in liver diseases and cancer. Int Immunopharmacol 2023; 120:110284. [PMID: 37196562 DOI: 10.1016/j.intimp.2023.110284] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
Exosomes are nanoscale (40-100 nm) vesicles secreted by different types of cells and have attracted extensive interest in recent years because of their unique role in disease development. It can carry related goods, such as lipids, proteins, and nucleic acids, to mediate intercellular communication. This review summarizes exosome biogenesis, release, uptake, and their role in mediating the development of liver diseases and cancer, such as viral hepatitis, drug-induced liver injury, alcohol-related liver disease, non-alcoholic fatty liver disease, hepatocellular carcinoma, and other tumors. Meanwhile, a fossa structural protein, caveolin-1(CAV-1), has also been proposed to be involved in the development of various diseases, especially liver diseases and tumors. In this review, we discuss the role of CAV-1 in liver diseases and different tumor stages (inhibition of early growth and promotion of late metastasis) and the underlying mechanisms by which CAV-1 regulates the process. In addition, CAV-1 has also been found to be a secreted protein that can be released directly through the exosome pathway or change the cargo composition of the exosomes, thus contributing to enhancing the metastasis and invasion of cancer cells during the late stage of tumor development. In conclusion, the role of CAV-1 and exosomes in disease development and the association between them remains to be one challenging uncharted area.
Collapse
Affiliation(s)
- Xiangfu Jiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Shuai Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Chengmu Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
72
|
Yamamoto A, Toba M, Takahashi Y, Takakura Y. Pharmacokinetic Approach for the Elucidation of Elevated Plasma Small Extracellular Vesicle (sEV) Concentration in Cancer. J Pharm Sci 2023; 112:1967-1974. [PMID: 37001861 DOI: 10.1016/j.xphs.2023.03.017] [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: 01/07/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023]
Abstract
The abundance of circulating plasma small extracellular vesicles (sEVs) has been reported to be elevated in cancer; however, the underlying mechanism remains unclear. In this study, a pharmacokinetic approach was used to determine the factors contributing to elevated plasma sEV levels during cancer in a tumor-bearing mouse model. Mouse plasma-derived sEVs (MP-sEVs) isolated from tumor-bearing mice showed increased protein concentrations and physicochemical characteristics comparable to MP-sEVs isolated from healthy mice. The steady-state concentration of sEVs is determined by the balance between the MP-sEV production and clearance. Thus, to determine whether tumorigenesis influences sEV clearance, isolated MP-sEVs were intravenously administered to either tumor-bearing or healthy mice. The results showed minimal differences in sEV clearance rates, suggesting that sEV production is the driving force of elevated MP-sEV concentrations. Lastly, CD63-gLuc stably expressing B16BL6-bearing mice were used to estimate the contribution of tumor cell-derived sEVs in the plasma. The gLuc activity of the MP-sEVs isolated was below the limit of detection, and it was estimated that the tumor cell-derived sEVs comprised at most 0.5% of the total MP-sEVs. Taken together, these results suggest that cells other than tumor cells contribute to elevated plasma sEV levels in cancer.
Collapse
Affiliation(s)
- Aki Yamamoto
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mihiro Toba
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
73
|
Mahmoodpour M, Kiasari BA, Karimi M, Abroshan A, Shamshirian D, Hosseinalizadeh H, Delavari A, Mirzei H. Paper-based biosensors as point-of-care diagnostic devices for the detection of cancers: a review of innovative techniques and clinical applications. Front Oncol 2023; 13:1131435. [PMID: 37456253 PMCID: PMC10348714 DOI: 10.3389/fonc.2023.1131435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
The development and rapid progression of cancer are major social problems. Medical diagnostic techniques and smooth clinical care of cancer are new necessities that must be supported by innovative diagnostic methods and technologies. Current molecular diagnostic tools based on the detection of blood protein markers are the most common tools for cancer diagnosis. Biosensors have already proven to be a cost-effective and accessible diagnostic tool that can be used where conventional laboratory methods are not readily available. Paper-based biosensors offer a new look at the world of analytical techniques by overcoming limitations through the creation of a simple device with significant advantages such as adaptability, biocompatibility, biodegradability, ease of use, large surface-to-volume ratio, and cost-effectiveness. In this review, we covered the characteristics of exosomes and their role in tumor growth and clinical diagnosis, followed by a discussion of various paper-based biosensors for exosome detection, such as dipsticks, lateral flow assays (LFA), and microfluidic paper-based devices (µPADs). We also discussed the various clinical studies on paper-based biosensors for exosome detection.
Collapse
Affiliation(s)
- Mehrdad Mahmoodpour
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Bahman Abedi Kiasari
- Virology Department, Faculty of Veterinary, The University of Tehran, Tehran, Iran
| | - Merat Karimi
- Institute of Nanoscience and Nanotechnology, University of Kashan, Kashan, Iran
| | - Arezou Abroshan
- Student Research Committee, Faculty of Veterinary Medicine, Shahid Bahonar University, Kerman, Iran
| | - Danial Shamshirian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alireza Delavari
- Student's Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
74
|
Zhang H, Zheng X, Zhao T, Chen Y, Luo Y, Dong Y, Tang H, Jiang J. Real-Time Monitoring of Exosomes Secretion from Single Cell Using Dual-Nanopore Biosensors. ACS Sens 2023. [PMID: 37368982 DOI: 10.1021/acssensors.3c00288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Exosomes secreted from cells carry rich information from their parent cells, representing a promising biomarker for investigation of diseases. We develop a dual-nanopore biosensor using DNA aptamers to specifically recognize CD63 protein on the exosome's surface, which enables label-free exosome detection based on ionic current change. The sensor allows for sensitive detection of exosomes with a detection limit of 3.4 × 106 particles/mL. The dual-nanopore biosensor was able to form an intrapipette electric circuit for ionic current measurement due to its unique structure, which is crucial to achieve detection of exosome secretion from a single cell. We utilized a microwell array chip to entrap a single cell into a confined microwell with small volume, enabling the accumulation of exosomes with high concentration. The dual-nanopore biosensor was positioned into the microwell with a single cell, and monitoring of exosome secretion from a single cell in different cell lines and under different stimulations has been achieved. Our design may provide a useful platform for developing nanopore biosensors for detecting cell secretions from a single living cell.
Collapse
Affiliation(s)
- Hongshuai Zhang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
- Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver Disease, Clinical Research Institute, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Xin Zheng
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tao Zhao
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yiping Chen
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yang Luo
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yangcan Dong
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Hao Tang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
75
|
Bhaoighill MN, Falcón‐Pérez JM, Royo F, Tee AR, Webber JP, Dunlop EA. Tuberous Sclerosis Complex cell-derived EVs have an altered protein cargo capable of regulating their microenvironment and have potential as disease biomarkers. J Extracell Vesicles 2023; 12:e12336. [PMID: 37337371 PMCID: PMC10279809 DOI: 10.1002/jev2.12336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/12/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
Hyperactivation of mechanistic target of rapamycin complex 1 (mTORC1) is a feature of many solid tumours and is a key pathogenic driver in the inherited condition Tuberous Sclerosis Complex (TSC). Modulation of the tumour microenvironment by extracellular vesicles (EVs) is known to facilitate the development of various cancers. The role of EVs in modulating the tumour microenvironment and their impact on the development of TSC tumours, however, remains unclear. This study, therefore, focuses on the poorly defined contribution of EVs to tumour growth in TSC. We characterised EVs secreted from TSC2-deficient and TSC2-expressing cells and identified a distinct protein cargo in TSC2-deficient EVs, containing an enrichment of proteins thought to be involved in tumour-supporting signalling pathways. We show EVs from TSC2-deficient cells promote cell viability, proliferation and growth factor secretion from recipient fibroblasts within the tumour microenvironment. Rapalogs (mTORC1 inhibitors) are the current therapy for TSC tumours. Here, we demonstrate a previously unknown intercellular therapeutic effect of rapamycin in altering EV cargo and reducing capacity to promote cell proliferation in the tumour microenvironment. Furthermore, EV cargo proteins have the potential for clinical applications as TSC biomarkers, and we reveal three EV-associated proteins that are elevated in plasma from TSC patients compared to healthy donor plasma.
Collapse
Affiliation(s)
- Muireann Ní Bhaoighill
- Tissue Microenvironment GroupSchool of MedicineCardiff UniversityCardiffUK
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Juan M. Falcón‐Pérez
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
- IKERBASQUEBasque Foundation for ScienceBilbaoSpain
| | - Félix Royo
- Exosomes Lab. CICbioGUNE‐BRTAParque TecnologicoDerioSpain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd)MadridSpain
| | - Andrew R. Tee
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| | - Jason P. Webber
- Tissue Microenvironment GroupSchool of MedicineCardiff UniversityCardiffUK
- Institute of Life ScienceSwansea University Medical SchoolSwansea UniversitySwanseaUK
| | - Elaine A. Dunlop
- Division of Cancer and GeneticsSchool of MedicineCardiff UniversityCardiffUK
| |
Collapse
|
76
|
Burlingham WJ, Jankowska-Gan E, Fechner JH, Little CJ, Wang J, Hong S, Molla M, Sullivan JA, Foley DP. Extracellular Vesicle-associated GARP/TGFβ:LAP Mediates "Infectious" Allo-tolerance. Transplant Direct 2023; 9:e1475. [PMID: 37250483 PMCID: PMC10212611 DOI: 10.1097/txd.0000000000001475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 05/31/2023] Open
Abstract
Here we test the hypothesis that, like CD81-associated "latent" IL35, the transforming growth factor (TGF)β:latency-associated peptide (LAP)/glycoprotein A repetitions predominant (GARP) complex was also tethered to small extracellular vesicles (sEVs), aka exosomes, produced by lymphocytes from allo-tolerized mice. Once these sEVs are taken up by conventional T cells, we also test whether TGFβ could be activated suppressing the local immune response. Methods C57BL/6 mice were tolerized by i.p. injection of CBA/J splenocytes followed by anti-CD40L/CD154 antibody treatment on days 0, 2, and 4. On day 35, spleen and lymph nodes were extracted and isolated lymphocytes were restimulated with sonicates of CBA splenocytes overnight. sEVs were extracted from culture supernatants by ultracentrifugation (100 000g) and assayed for (a) the presence of TGFβ:LAP associated with tetraspanins CD81,CD63, and CD9 by enzyme-linked immunosorbent assay; (b) GARP, critical to membrane association of TGFβ:LAP and to activation from its latent form, as well as various TGFβ receptors; and (c) TGFβ-dependent function in 1° and 2° immunosuppression of tetanus toxoid-immunized B6 splenocytes using trans-vivo delayed-type hypersensitivity assay. Results After tolerization, CBA-restimulated lymphocytes secreted GARP/TGFβ:LAP-coated extracellular vesicles. Like IL35 subunits, but unlike IL10, which was absent from ultracentrifuge pellets, GARP/TGFβ:LAP was mainly associated with CD81+ exosomes. sEV-bound GARP/TGFβ:LAP became active in both 1° and 2° immunosuppression, the latter requiring sEV uptake by "bystander" T cells and reexpression on the cell surface. Conclusions Like other immune-suppressive components of the Treg exosome, which are produced in a latent form, exosomal GARP/TGFβ:LAP produced by allo-specific regulatory T cells undergoes either immediate activation (1° suppression) or internalization by naive T cells, followed by surface reexpression and subsequent activation (2°), to become suppressive. Our results imply a membrane-associated form of TGFβ:LAP that, like exosomal IL35, can target "bystander" lymphocytes. This new finding implicates exosomal TGFβ:LAP along with Treg-derived GARP as part of the infectious tolerance network.
Collapse
Affiliation(s)
- William J. Burlingham
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Ewa Jankowska-Gan
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - John H. Fechner
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Christopher J. Little
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI
| | - Seungpyo Hong
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI
| | - Miraf Molla
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Jeremy A. Sullivan
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - David P. Foley
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
77
|
Logozzi M, Orefice NS, Di Raimo R, Mizzoni D, Fais S. The Importance of Detecting, Quantifying, and Characterizing Exosomes as a New Diagnostic/Prognostic Approach for Tumor Patients. Cancers (Basel) 2023; 15:cancers15112878. [PMID: 37296842 DOI: 10.3390/cancers15112878] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Exosomes are extracellular vesicles (EVs) of nanometric size studied for their role in tumor pathogenesis and progression and as a new source of tumor biomarkers. The clinical studies have provided encouraging but probably unexpected results, including the exosome plasmatic levels' clinical relevance and well-known biomarkers' overexpression on the circulating EVs. The technical approach to obtaining EVs includes methods to physically purify EVs and characterize EVs, such as Nanosight Tracking Analysis (NTA), immunocapture-based ELISA, and nano-scale flow cytometry. Based on the above approaches, some clinical investigations have been performed on patients with different tumors, providing exciting and promising results. Here we emphasize data showing that exosome plasmatic levels are consistently higher in tumor patients than in controls and that plasmatic exosomes express well-known tumor markers (e.g., PSA and CEA), proteins with enzymatic activity, and nucleic acids. However, we also know that tumor microenvironment acidity is a key factor in influencing both the amount and the characteristics of the exosome released by tumor cells. In fact, acidity significantly increases exosome release by tumor cells, which correlates with the number of exosomes that circulate through the body of a tumor patient.
Collapse
Affiliation(s)
- Mariantonia Logozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Nicola Salvatore Orefice
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | - Davide Mizzoni
- ExoLab Italia, Tecnopolo d'Abruzzo, 67100 L'Aquila, Italy
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
78
|
Li Z, Gao Y, Cao Y, He F, Jiang R, Liu H, Cai H, Zan T. Extracellular RNA in melanoma: Advances, challenges, and opportunities. Front Cell Dev Biol 2023; 11:1141543. [PMID: 37215082 PMCID: PMC10192583 DOI: 10.3389/fcell.2023.1141543] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Melanoma, a malignant mass lesion that originates in melanocytes and has a high rate of malignancy, metastasis, and mortality, is defined by these characteristics. Malignant melanoma is a kind of highly malignant tumor that produces melanin and has a high mortality rate. Its incidence accounts for 1%-3% of all malignant tumors and shows an obvious upward trend. The discovery of biomolecules for the diagnosis and treatment of malignant melanoma has important application value. So far, the exact molecular mechanism of melanoma development relevant signal pathway still remains unclear. According to previous studies, extracellular RNAs (exRNAs) have been implicated in tumorigenesis and spread of melanoma. They can influence the proliferation, invasion and metastasis of melanoma by controlling the expression of target genes and can also influence tumor progression by participating in signal transduction mechanisms. Therefore, understanding the relationship between exRNA and malignant melanoma and targeting therapy is of positive significance for its prevention and treatment. In this review, we did an analysis of extracellular vesicles of melanoma which focused on the role of exRNAs (lncRNAs, miRNAs, and mRNAs) and identifies several potential therapeutic targets. In addition, we discuss the typical signaling pathways involved in exRNAs, advances in exRNA detection and how they affect the tumor immune microenvironment in melanoma.
Collapse
Affiliation(s)
- Zhouxiao Li
- Department of Plastic and Reconstructive Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Gao
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Cao
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feifan He
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Runyi Jiang
- Department of Orthopaedic Oncology, Spinal Tumor Center, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hanyuan Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
79
|
Ko SY, Lee W, Weigert M, Jonasch E, Lengyel E, Naora H. The glycoprotein CD147 defines miRNA-enriched extracellular vesicles that derive from cancer cells. J Extracell Vesicles 2023; 12:e12318. [PMID: 36973758 PMCID: PMC10042814 DOI: 10.1002/jev2.12318] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Extracellular vesicles (EVs) are ideal for liquid biopsy, but distinguishing cancer cell-derived EVs and subpopulations of biomarker-containing EVs in body fluids has been challenging. Here, we identified that the glycoproteins CD147 and CD98 define subpopulations of EVs that are distinct from classical tetraspanin+ EVs in their biogenesis. Notably, we identified that CD147+ EVs have substantially higher microRNA (miRNA) content than tetraspanin+ EVs and are selectively enriched in miRNA through the interaction of CD147 with heterogeneous nuclear ribonucleoprotein A2/B1. Studies using mouse xenograft models showed that CD147+ EVs predominantly derive from cancer cells, whereas the majority of tetraspanin+ EVs are not of cancer cell origin. Circulating CD147+ EVs, but not tetraspanin+ EVs, were significantly increased in prevalence in patients with ovarian and renal cancers as compared to healthy individuals and patients with benign conditions. Furthermore, we found that isolating miRNAs from body fluids by CD147 immunocapture increases the sensitivity of detecting cancer cell-specific miRNAs, and that circulating miRNAs isolated by CD147 immunocapture more closely reflect the tumor miRNA signature than circulating miRNAs isolated by conventional methods. Collectively, our findings reveal that CD147 defines miRNA-enriched, cancer cell-derived EVs, and that CD147 immunocapture could be an effective approach to isolate cancer-derived miRNAs for liquid biopsy.
Collapse
Affiliation(s)
- Song Yi Ko
- Department of Molecular and Cellular OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - WonJae Lee
- Department of Molecular and Cellular OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Melanie Weigert
- Section of Gynecologic OncologyDepartment of Obstetrics and GynecologyUniversity of ChicagoChicagoIllinoisUSA
| | - Eric Jonasch
- Department of Genitourinary Medical OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Ernst Lengyel
- Section of Gynecologic OncologyDepartment of Obstetrics and GynecologyUniversity of ChicagoChicagoIllinoisUSA
| | - Honami Naora
- Department of Molecular and Cellular OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
80
|
Acevedo-Sánchez V, Martínez-Ruiz RS, Aguilar-Ruíz SR, Torres-Aguilar H, Chávez-Olmos P, Garrido E, Baltiérrez-Hoyos R, Romero-Tlalolini MDLA. Quantitative Proteomics for the Identification of Differentially Expressed Proteins in the Extracellular Vesicles of Cervical Cancer Cells. Viruses 2023; 15:702. [PMID: 36992411 PMCID: PMC10051161 DOI: 10.3390/v15030702] [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: 02/04/2023] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The extracellular vesicles (EVs) in a tumoral microenvironment can exert different functions by transferring their content, which has been poorly described in cervical cancer. Here, we tried to clarify the proteomic content of these EVs, comparing those derived from cancerous HPV (+) keratinocytes (HeLa) versus those derived from normal HPV (-) keratinocytes (HaCaT). We performed a quantitative proteomic analysis, using LC-MS/MS, of the EVs from HeLa and HaCaT cell lines. The up- and downregulated proteins in the EVs from the HeLa cell line were established, along with the cellular component, molecular function, biological processes, and signaling pathways in which they participate. The biological processes with the highest number of upregulated proteins are cell adhesion, proteolysis, lipid metabolic process, and immune system processes. Interestingly, three of the top five signaling pathways with more up- and downregulated proteins are part of the immune response. Due to their content, we can infer that EVs can have a significant role in migration, invasion, metastasis, and the activation or suppression of immune system cells in cancer.
Collapse
Affiliation(s)
- Víctor Acevedo-Sánchez
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez 68120, Mexico
| | - Roy S. Martínez-Ruiz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez 68120, Mexico
| | - Sergio R. Aguilar-Ruíz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez 68120, Mexico
| | - Honorio Torres-Aguilar
- Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Av. Universidad S/N, Cinco Señores, Oaxaca de Juárez 68120, Mexico
| | - Pedro Chávez-Olmos
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación Gustavo A. Madero, Mexico City 07360, Mexico
| | - Efraín Garrido
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación Gustavo A. Madero, Mexico City 07360, Mexico
| | - Rafael Baltiérrez-Hoyos
- Facultad de Medicina y Cirugía, CONACYT—Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez 68120, Mexico
| | - María de los A. Romero-Tlalolini
- Facultad de Medicina y Cirugía, CONACYT—Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez 68120, Mexico
| |
Collapse
|
81
|
Malgundkar SH, Tamimi Y. Exosomes as crucial emerging tools for intercellular communication with therapeutic potential in ovarian cancer. Future Sci OA 2023; 9:FSO833. [PMID: 37006229 PMCID: PMC10051132 DOI: 10.2144/fsoa-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than two-thirds of epithelial ovarian cancer (EOC) patients are diagnosed at advanced stages due to the lack of sensitive biomarkers. Currently, exosomes are intensively investigated as non-invasive cancer diagnostic markers. Exosomes are nanovesicles released in the extracellular milieu with the potential to modulate recipient cells' behavior. EOC cells release many altered exosomal cargoes that exhibit clinical relevance to tumor progression. Exosomes represent powerful therapeutic tools (drug carriers or vaccines), posing a promising option in clinical practice for curing EOC in the near future. In this review, we highlight the importance of exosomes in cell–cell communication, epithelial–mesenchymal transition (EMT), and their potential to serve as diagnostic and prognostic factors, particularly in EOC.
Collapse
Affiliation(s)
- Shika Hanif Malgundkar
- Department of Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, PO Box 35, PC 123, Muscat, Sultanate of Oman
| | - Yahya Tamimi
- Department of Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, PO Box 35, PC 123, Muscat, Sultanate of Oman
| |
Collapse
|
82
|
Magoling BJA, Wu AYT, Chen YJ, Wong WWT, Chuo STY, Huang HC, Sung YC, Hsieh HT, Huang P, Lee KZ, Huang KW, Chen RH, Chen Y, Lai CPK. Membrane Protein Modification Modulates Big and Small Extracellular Vesicle Biodistribution and Tumorigenic Potential in Breast Cancers In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208966. [PMID: 36609913 DOI: 10.1002/adma.202208966] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) are released by cells to mediate intercellular communication under pathological and physiological conditions. While small EVs (sEVs; <100-200 nm, exosomes) are intensely investigated, the properties and functions of medium and large EVs (big EVs (bEVs); >200 nm, microvesicles) are less well explored. Here, bEVs and sEVs are identified as distinct EV populations, and it is determined that bEVs are released in a greater bEV:sEV ratio in the aggressive human triple-negative breast cancer (TNBC) subtype. PalmGRET, bioluminescence-resonance-energy-transfer (BRET)-based EV reporter, reveals dose-dependent EV biodistribution at nonlethal and physiological EV dosages, as compared to lipophilic fluorescent dyes. Remarkably, the bEVs and sEVs exhibit unique biodistribution profiles, yet individually promote in vivo tumor growth in a syngeneic immunocompetent TNBC breast tumor murine model. The bEVs and sEVs share mass-spectrometry-identified tumor-progression-associated EV surface membrane proteins (tpEVSurfMEMs), which include solute carrier family 29 member 1, Cd9, and Cd44. tpEVSurfMEM depletion attenuates EV lung organotropism, alters biodistribution, and reduces protumorigenic potential. This study identifies distinct in vivo property and function of bEVs and sEVs in breast cancer, which suggest the significant role of bEVs in diseases, diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Bryan John Abel Magoling
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
| | - Anthony Yan-Tang Wu
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yen-Ju Chen
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Wendy Wan-Ting Wong
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Steven Ting-Yu Chuo
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Hsi-Chien Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hsin Tzu Hsieh
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Poya Huang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Kang-Zhang Lee
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
| | - Kuan-Wei Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ruey-Hwa Chen
- Graduate Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 10617, Taiwan
- Chemical Biology and Molecular Biophysics Program, TIGP, Academia Sinica, Taipei, 11529, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
83
|
Exosome-Transmitted miR-506-3p Inhibits Colorectal Cancer Cell Malignancy via Regulating GSTP1. Appl Biochem Biotechnol 2023; 195:2015-2027. [PMID: 36401721 DOI: 10.1007/s12010-022-04268-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/20/2022]
Abstract
Exosome-mediated microRNA transfer has been shown to regulate cancer progression. However, the involvement of exosomal-miR-506-3p in colorectal cancer (CRC) is unknown. The goal of the research was to study into the role of exosomal-miR-506-3p in CRC. Using a qRT-PCR experiment, it was observed that CRC tissues had lower levels of miR-506-3p than non-tumor tissues. It was observed that miR-506-3p inhibited the proliferation, regulates apoptosis, and cell cycle of HT29 and SW480 cells as compared to control groups. Dual luciferase reporter assay results showed that GSTP1 was the downstream target molecule of miR-506-3p, which was consistent with the database prediction. Furthermore, FHC cells transfected with miR-506-3p could transfer miR-506-3p to SW480 cells, limiting cell growth and inducing cell death. We discovered a unique regulatory mechanism in which exosome-mediated transfer of miR-506-3p reduces proliferation and induces apoptosis in CRC through negative regulation of GSTP1, implying that exosome-mediated delivery of miR-506-3p provides fresh insight into CRC diagnostics and treatment.
Collapse
|
84
|
Qiao L, Dong C, Zhang J, Sun G. The expression of Rab5 and its effect on invasion, migration and exosome secretion in triple negative breast cancer. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:157-165. [PMID: 36815255 PMCID: PMC9968947 DOI: 10.4196/kjpp.2023.27.2.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 02/24/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and current therapeutic strategies are limited in their effectiveness. The expressions of Rab5 and the M2 tumor-associated macrophage marker CD163 in tissues were detected by Western blot. The migration and invasion of cells were determined using a Transwell assay. The expressions of the exosome markers were evaluated by Western blot. The polarization of human macrophages (THP-1) was determined by incubation of THP-1 cells with conditioned medium or exosomes collected from MDA-MB-231 cells with indicated transfections or by a coculture system of THP-1 and MDA-MB-231 cells. The M1 and M2 macrophage markers were evaluated by qRT-PCR. The expression of Rab5 in TNBC was significantly higher than that in normal breast tissue. Rab5 expressions in triple-negative and luminal A breast cancer were higher than those in other molecular subtypes. Higher CD163 expression was observed in triple-negative breast cancer and in triple-negative and luminal B subtypes. Rab5 knockdown suppressed but Rab5 overexpression promoted the migration and invasion capacity of MDA-MB-231 cells. The levels of CD63 and CD9 in the medium of Rab5 knockdown cells were lower than those in control cells, whereas higher levels of CD63 and CD9 were observed in Rab5 overexpression cells. Rab5 knockdown decreased the excretion but did not alter the diameter of the exosomes. Knockdown of Rab5 facilitated the anti-tumor polarization of macrophages, which was partially reversed by Rab5 overexpression. Therefore, Rab5 is expected to be a potential therapeutic target for triple-negative breast cancer.
Collapse
Affiliation(s)
- Lei Qiao
- Departments of Breast and Thyroid Surgery, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang Province 830000, China
| | - Chao Dong
- Departments of Breast and Thyroid Surgery, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang Province 830000, China
| | - Jiaojiao Zhang
- Departments of Anesthesia and Perioperative Medicine, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang Province 830000, China
| | - Gang Sun
- Departments of Breast and Thyroid Surgery, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang Province 830000, China,Correspondence Gang Sun, E-mail:
| |
Collapse
|
85
|
A review on comparative studies addressing exosome isolation methods from body fluids. Anal Bioanal Chem 2023; 415:1239-1263. [PMID: 35838769 DOI: 10.1007/s00216-022-04174-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/10/2022] [Indexed: 12/11/2022]
Abstract
Exosomes emerged as valuable sources of disease biomarkers and new therapeutic tools. However, extracellular vesicles isolation with exosome-like characteristics from certain biofluids is still challenging which can limit their potential use in clinical settings. While ultracentrifugation-based procedures are the gold standard for exosome isolation from cell cultures, no unique and standardized method for exosome isolation from distinct body fluids exists. The complexity, specific composition, and physical properties of each biofluid constitute a technical barrier to obtain reproducible and pure exosome preparations, demanding a detailed characterization of both exosome isolation and characterization methods. Moreover, some isolation procedures can affect downstream proteomic or RNA profiling analysis. This review compiles and discussed a set of comparative studies addressing distinct exosome isolation methods from human biofluids, including cerebrospinal fluid, plasma, serum, saliva, and urine, also focusing on body fluid specific challenges, physical properties, and other potential variation sources. This summarized information will facilitate the choice of exosome isolation methods, based on the type of biological samples available, and hopefully encourage the use of exosomes in translational and clinical research.
Collapse
|
86
|
Jiang B, Xie D, Wang S, Li X, Wu G. Advances in early detection methods for solid tumors. Front Genet 2023; 14:1091223. [PMID: 36911396 PMCID: PMC9998680 DOI: 10.3389/fgene.2023.1091223] [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: 11/06/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
During the last decade, non-invasive methods such as liquid biopsy have slowly replaced traditional imaging and invasive pathological methods used to diagnose and monitor cancer. Improvements in the available detection methods have enabled the early screening and diagnosis of solid tumors. In addition, advances in early detection methods have made the continuous monitoring of tumor progression using repeat sampling possible. Previously, the focus of liquid biopsy techniques included the following: 1) the isolation of circulating tumor cells, circulating tumor DNA, and extracellular tumor vesicles from solid tumor cells in the patient's blood; in addition to 2) analyzing genomic and proteomic data contained within the isolates. Recently, there has been a rapid devolvement in the techniques used to isolate and analyze molecular markers. This rapid evolvement in detection techniques improves their accuracy, especially when few samples are available. In addition, there is a tremendous expansion in the acquisition of samples and targets for testing; solid tumors can be detected from blood and other body fluids. Test objects have also expanded from samples taken directly from cancer to include indirect objects affected in cancer development. Liquid biopsy technology has limitations. Even so, this detection technique is the key to a new phase of oncogenetics. This review aims to provide an overview of the current advances in liquid biopsy marker selection, isolation, and detection methods for solid tumors. The advantages and disadvantages of liquid biopsy technology will also be explored.
Collapse
Affiliation(s)
| | | | | | - Xiunan Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
87
|
Chattrairat K, Yasui T, Suzuki S, Natsume A, Nagashima K, Iida M, Zhang M, Shimada T, Kato A, Aoki K, Ohka F, Yamazaki S, Yanagida T, Baba Y. All-in-One Nanowire Assay System for Capture and Analysis of Extracellular Vesicles from an ex Vivo Brain Tumor Model. ACS NANO 2023; 17:2235-2244. [PMID: 36655866 PMCID: PMC9933609 DOI: 10.1021/acsnano.2c08526] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/04/2023] [Indexed: 05/27/2023]
Abstract
Extracellular vesicles (EVs) have promising potential as biomarkers for early cancer diagnosis. The EVs have been widely studied as biological cargo containing essential biological information not only from inside vesicles such as nucleic acids and proteins but also from outside vesicles such as membrane proteins and glycolipids. Although various methods have been developed to isolate EVs with high yields such as captures based on density, size, and immunoaffinity, different measurement systems are needed to analyze EVs after isolation, and a platform that enables all-in-one analysis of EVs from capture to detection in multiple samples is desired. Since a nanowire-based approach has shown an effective capability for capturing EVs via surface charge interaction compared to other conventional methods, here, we upgraded the conventional well plate assay to an all-in-one nanowire-integrated well plate assay system (i.e., a nanowire assay system) that enables charge-based EV capture and EV analysis of membrane proteins. We applied the nanowire assay system to analyze EVs from brain tumor organoids in which tumor environments, including vascular formations, were reconstructed, and we found that the membrane protein expression ratio of CD31/CD63 was 1.42-fold higher in the tumor organoid-derived EVs with a p-value less than 0.05. Furthermore, this ratio for urine samples from glioblastoma patients was 2.25-fold higher than that from noncancer subjects with a p-value less than 0.05 as well. Our results demonstrated that the conventional well plate method integrated with the nanowire-based EV capture approach allows users not only to capture EVs effectively but also to analyze them in one assay system. We anticipate that the all-in-one nanowire assay system will be a powerful tool for elucidating EV-mediated tumor-microenvironment crosstalk.
Collapse
Affiliation(s)
- Kunanon Chattrairat
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Takao Yasui
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Japan
Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Shunsuke Suzuki
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Atsushi Natsume
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kazuki Nagashima
- Japan
Science and Technology Agency (JST), PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Department
of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Mikiko Iida
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Min Zhang
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Taisuke Shimada
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Akira Kato
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kosuke Aoki
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Fumiharu Ohka
- Department
of Neurosurgery, School of Medicine, Nagoya
University, 65 Tsurumai-cho,
Showa-ku, Nagoya 466-8550, Japan
| | - Shintaro Yamazaki
- Department
of Neurosurgery, School of Medicine, Nagoya
University, 65 Tsurumai-cho,
Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Yanagida
- Department
of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yoshinobu Baba
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute
of Quantum Life Science, National Institutes
for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
88
|
Surman M, Jankowska U, Wilczak M, Przybyło M. Similarities and Differences in the Protein Composition of Cutaneous Melanoma Cells and Their Exosomes Identified by Mass Spectrometry. Cancers (Basel) 2023; 15:cancers15041097. [PMID: 36831440 PMCID: PMC9954195 DOI: 10.3390/cancers15041097] [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: 12/21/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Intercellular transport of proteins mediated by extracellular vesicles (EVs)-exosomes and ectosomes-is one of the factors facilitating carcinogenesis. Therefore, the research on protein cargo of melanoma-derived EVs may provide a better understanding of the mechanisms involved in melanoma progression and contribute to the development of alternative biomarkers. Proteomic data on melanoma-derived EVs are very limited. The shotgun nanoLC-MS/MS approach was applied to analyze the protein composition of primary (WM115, WM793) and metastatic (WM266-4, WM1205Lu) cutaneous melanoma cells and exosomes released by them. All cells secreted homogeneous populations of exosomes that shared a characteristic set of proteins. In total, 3514 and 1234 unique proteins were identified in melanoma cells and exosomes, respectively. Gene ontology analysis showed enrichment in several cancer-related categories, including cell proliferation, migration, negative regulation of apoptosis, and angiogenesis. The obtained results broaden our knowledge on the role of selected proteins in exosome biology, as well as their functional role in the development and progression of cutaneous melanoma. The results may also inspire future studies on the clinical potential of exosomes.
Collapse
Affiliation(s)
- Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Correspondence: ; Tel.: +48-12-664-6462
| |
Collapse
|
89
|
Cardiovascular Disease as a Consequence or a Cause of Cancer: Potential Role of Extracellular Vesicles. Biomolecules 2023; 13:biom13020321. [PMID: 36830690 PMCID: PMC9953640 DOI: 10.3390/biom13020321] [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: 12/07/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Both cardiovascular disease and cancer continue to be causes of morbidity and mortality all over the world. Preventing and treating heart disease in patients undergoing cancer treatment remain an important and ongoing challenge for improving the lives of cancer patients, but also for their survival. Despite ongoing efforts to improve patient survival, minimal advances have been made in the early detection of cardiovascular disease in patients suffering from cancer. Understanding the communication between cancer and cardiovascular disease can be based on a deeper knowledge of the molecular mechanisms that define the profile of the bilateral network and establish disease-specific biomarkers and therapeutic targets. The role of exosomes, microvesicles, and apoptotic bodies, together defined as extracellular vesicles (EVs), in cross talk between cardiovascular disease and cancer is in an incipient form of research. Here, we will discuss the preclinical evidence on the bilateral connection between cancer and cardiovascular disease (especially early cardiac changes) through some specific mediators such as EVs. Investigating EV-based biomarkers and therapies may uncover the responsible mechanisms, detect the early stages of cardiovascular damage and elucidate novel therapeutic approaches. The ultimate goal is to reduce the burden of cardiovascular diseases by improving the standard of care in oncological patients treated with anticancer drugs or radiotherapy.
Collapse
|
90
|
Musi A, Bongiovanni L. Extracellular Vesicles in Cancer Drug Resistance: Implications on Melanoma Therapy. Cancers (Basel) 2023; 15:1074. [PMID: 36831417 PMCID: PMC9954626 DOI: 10.3390/cancers15041074] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Extracellular vesicles (EVs) are involved in the pathogenesis of neoplastic diseases. Their role in mediating drug resistance has been widely described in several types of cancers, including melanoma. EVs can mediate drug resistance through several different mechanisms, such as drug-sequestration, transfer of pro-survival proteins and RNA, induction of cancer stem cell-like features and interaction with cells of the tumor microenvironment and immune-system. Melanoma is a highly immunogenic tumor originating from the malignant transformation of melanocytes. Several therapeutic strategies currently used in the treatment of melanoma and the combination of BRAF and MEK-inhibitors, as well as immune check-point inhibitors (ICI), have consistently improved the overall survival time of melanoma patients. However, the development of resistance is one of the biggest problems leading to a poor clinical outcome, and EVs can contribute to this. EVs isolated from melanoma cells can contain "sequestered" chemotherapeutic drugs in order to eliminate them, or bioactive molecules (such as miRNA or proteins) that have been proven to play a crucial role in the transmission of resistance to sensitive neoplastic cells. This leads to the hypothesis that EVs could be considered as resistance-mediators in sensitive melanoma cells. These findings are a pivotal starting point for further investigations to better understand EVs' role in drug resistance mechanisms and how to target them. The purpose of this review is to summarize knowledge about EVs in order to develop a deeper understanding of their underlying mechanisms. This could lead to the development of new therapeutic strategies able to bypass EV-mediated drug-resistance in melanoma, such as by the use of combination therapy, including EV release inhibitors.
Collapse
Affiliation(s)
- Alice Musi
- Department of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
| | - Laura Bongiovanni
- Department of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CT Utrecht, The Netherlands
| |
Collapse
|
91
|
Bajo-Santos C, Brokāne A, Zayakin P, Endzeliņš E, Soboļevska K, Belovs A, Jansons J, Sperga M, Llorente A, Radoviča-Spalviņa I, Lietuvietis V, Linē A. Plasma and urinary extracellular vesicles as a source of RNA biomarkers for prostate cancer in liquid biopsies. Front Mol Biosci 2023; 10:980433. [PMID: 36818049 PMCID: PMC9935579 DOI: 10.3389/fmolb.2023.980433] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Extracellular vesicles (EVs) have emerged as a very attractive source of cancer- derived RNA biomarkers for the early detection, prognosis and monitoring of various cancers, including prostate cancer (PC). However, biofluids contain a mixture of EVs released from a variety of tissues and the fraction of total EVs that are derived from PC tissue is not known. Moreover, the optimal biofluid-plasma or urine-that is more suitable for the detection of EV- enclosed RNA biomarkers is not yet clear. Methodology: In the current study, we performed RNA sequencing analysis of plasma and urinary EVs collected before and after radical prostatectomy, and matched tumor and normal prostate tissues of 10 patients with prostate cancer. Results and Discussion: The most abundant RNA biotypes in EVs were miRNA, piRNA, tRNA, lncRNA, rRNA and mRNA. To identify putative cancer-derived RNA biomarkers, we searched for RNAs that were overexpressed in tumor as compared to normal tissues, present in the pre-operation EVs and decreased in the post-operation EVs in each RNA biotype. The levels of 63 mRNAs, 3 lncRNAs, 2 miRNAs and 1 piRNA were significantly increased in the tumors and decreased in the post-operation urinary EVs, thus suggesting that these RNAs mainly originate from PC tissue. No such RNA biomarkers were identified in plasma EVs. This suggests that the fraction of PC-derived EVs in urine is larger than in plasma and allows the detection and tracking of PC-derived RNAs.
Collapse
Affiliation(s)
| | - Agnese Brokāne
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Pawel Zayakin
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | | | | | | | - Alicia Llorente
- Department Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| | | | | | - Aija Linē
- Latvian Biomedical Research and Study Centre, Riga, Latvia,*Correspondence: Aija Linē,
| |
Collapse
|
92
|
Rangel-Ramírez VV, González-Sánchez HM, Lucio-García C. Exosomes: from biology to immunotherapy in infectious diseases. Infect Dis (Lond) 2023; 55:79-107. [PMID: 36562253 DOI: 10.1080/23744235.2022.2149852] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Exosomes are extracellular vesicles derived from the endosomal compartment, which are released by all kinds of eukaryotic and prokaryotic organisms. These vesicles contain a variety of biomolecules that differ both in quantity and type depending on the origin and cellular state. Exosomes are internalized by recipient cells, delivering their content and thus contributing to cell-cell communication in health and disease. During infections exosomes may exert a dual role, on one hand, they can transmit pathogen-related molecules mediating further infection and damage, and on the other hand, they can protect the host by activating the immune response and reducing pathogen spread. Selective packaging of pathogenic components may mediate these effects. Recently, quantitative analysis of samples by omics technologies has allowed a deep characterization of the proteins, lipids, RNA, and metabolite cargoes of exosomes. Knowledge about the content of these vesicles may facilitate their therapeutic application. Furthermore, as exosomes have been detected in almost all biological fluids, pathogenic or host-derived components can be identified in liquid biopsies, making them suitable for diagnosis and prognosis. This review attempts to organize the recent findings on exosome composition and function during viral, bacterial, fungal, and protozoan infections, and their contribution to host defense or to pathogen spread. Moreover, we summarize the current perspectives and future directions regarding the potential application of exosomes for prophylactic and therapeutic purposes.
Collapse
Affiliation(s)
| | | | - César Lucio-García
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, México
| |
Collapse
|
93
|
Medhin LB, Beasley AB, Warburton L, Amanuel B, Gray ES. Extracellular vesicles as a liquid biopsy for melanoma: Are we there yet? Semin Cancer Biol 2023; 89:92-98. [PMID: 36706847 DOI: 10.1016/j.semcancer.2023.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Melanoma is the most aggressive form of skin cancer owing to its high propensity to metastasise in distant organs and develop resistance to treatment. The scarce treatment options available for melanoma underscore the need for biomarkers to guide treatment decisions. In this context, an attractive alternative to overcome the limitations of repeated tissue sampling is the analysis of peripheral blood samples, referred to as 'liquid biopsy'. In particular, the analysis of extracellular vesicles (EVs) has emerged as a promising candidate due to their role in orchestrating cancer dissemination, immune modulation, and drug resistance. As we gain insights into the role of EVs in cancer and melanoma their potential for clinical use is becoming apparent. Herein, we critically summarise the current evidence supporting EVs as biomarkers for melanoma diagnosis, prognostication, therapy response prediction, and drug resistance. EVs are proposed as a candidate biomarker for predicting therapeutic response to immune checkpoint inhibition. However, to realise the potential of EV analysis for clinical decision-making strong clinical validation is required, underscoring the need for further research in this area.
Collapse
Affiliation(s)
- Lidia B Medhin
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia
| | - Aaron B Beasley
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia
| | - Lydia Warburton
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia; Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia
| | - Benhur Amanuel
- School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia; Department of Anatomical Pathology PathWest, QEII Medical Centre, Nedlands WA 6009, Australia
| | - Elin S Gray
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia.
| |
Collapse
|
94
|
Peruzzu D, Boussadia Z, Fratini F, Spadaro F, Bertuccini L, Sanchez M, Carollo M, Matarrese P, Falchi M, Iosi F, Raggi C, Parolini I, Carè A, Sargiacomo M, Gagliardi MC, Fecchi K. Inhibition of cholesterol transport impairs Cav-1 trafficking and small extracellular vesicles secretion, promoting amphisome formation in melanoma cells. Traffic 2023; 24:76-94. [PMID: 36519961 DOI: 10.1111/tra.12878] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Caveolin-1 (Cav-1) is a fundamental constituent of caveolae, whose functionality and structure are strictly dependent on cholesterol. In this work the U18666A inhibitor was used to study the role of cholesterol transport in the endosomal degradative-secretory system in a metastatic human melanoma cell line (WM266-4). We found that U18666A induces a shift of Cav-1 from the plasma membrane to the endolysosomal compartment, which is involved, through Multi Vesicular Bodies (MVBs), in the formation and release of small extracellular vesicles (sEVs). Moreover, this inhibitor induces an increase in the production of sEVs with chemical-physical characteristics similar to control sEVs but with a different protein composition (lower expression of Cav-1 and increase of LC3II) and reduced transfer capacity on target cells. Furthermore, we determined that U18666A affects mitochondrial function and also cancer cell aggressive features, such as migration and invasion. Taken together, these results indicate that the blockage of cholesterol transport, determining the internalization of Cav-1, may modify sEVs secretory pathways through an increased fusion between autophagosomes and MVBs to form amphisome, which in turn fuses with the plasma membrane releasing a heterogeneous population of sEVs to maintain homeostasis and ensure correct cellular functionality.
Collapse
Affiliation(s)
- Daniela Peruzzu
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Federica Fratini
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Spadaro
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Bertuccini
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Sanchez
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Carollo
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National Center for HIV/AIDS Research, Istituto Superiore di Sanità, Rome
| | - Francesca Iosi
- Core Facilities Technical Scientific Service, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Raggi
- National Center for the control and evaluation of Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Isabella Parolini
- Department Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Sargiacomo
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Katia Fecchi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
95
|
Stefańska K, Józkowiak M, Angelova Volponi A, Shibli JA, Golkar-Narenji A, Antosik P, Bukowska D, Piotrowska-Kempisty H, Mozdziak P, Dzięgiel P, Podhorska-Okołów M, Zabel M, Dyszkiewicz-Konwińska M, Kempisty B. The Role of Exosomes in Human Carcinogenesis and Cancer Therapy-Recent Findings from Molecular and Clinical Research. Cells 2023; 12:cells12030356. [PMID: 36766698 PMCID: PMC9913699 DOI: 10.3390/cells12030356] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Exosomes are biological nanoscale spherical lipid bilayer vesicles, 40-160 nm in diameter, produced by most mammalian cells in both physiological and pathological conditions. Exosomes are formed via the endosomal sorting complex required for transport (ESCRT). The primary function of exosomes is mediating cell-to-cell communication. In terms of cancer, exosomes play important roles as mediators of intercellular communication, leading to tumor progression. Moreover, they can serve as biomarkers for cancer detection and progression. Therefore, their utilization in cancer therapies has been suggested, either as drug delivery carriers or as a diagnostic tool. However, exosomes were also reported to be involved in cancer drug resistance via transferring information of drug resistance to sensitive cells. It is important to consider the current knowledge regarding the role of exosomes in cancer, drug resistance, cancer therapies, and their clinical application in cancer therapies.
Collapse
Affiliation(s)
- Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Cellivia 3 S.A., 61-623 Poznan, Poland
| | - Małgorzata Józkowiak
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Ana Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, Dental Institute, King’s College London, London WC2R 2LS, UK
| | - Jamil Awad Shibli
- Department of Periodontology and Oral Implantology, University of Guarulhos, Guarulhos 07030-010, Brazil
| | - Afsaneh Golkar-Narenji
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Marta Dyszkiewicz-Konwińska
- Department of Biomaterials and Experimental Dentistry, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Bartosz Kempisty
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic
- Correspondence:
| |
Collapse
|
96
|
Huang T, Sato Y, Kuramochi A, Ohba Y, Sano M, Miyagishi M, Tateno H, Wadhwa R, Kawasaki K, Uchida T, Ekdahl KN, Nilsson B, Chung UI, Teramura Y. Surface modulation of extracellular vesicles with cell-penetrating peptide-conjugated lipids for improvement of intracellular delivery to endothelial cells. Regen Ther 2023; 22:90-98. [PMID: 36712957 PMCID: PMC9842955 DOI: 10.1016/j.reth.2022.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Exosomes (diameter 30-200 nm) are a subtype of extracellular vesicles secreted by cells containing DNA, microRNA (miRNA), and proteins. Exosomes are expected to be valuable as a means of delivering drugs or functional miRNAs in treatment of diseases. However, the delivery of exosomes is not sufficiently effective, even though exosomes have intrinsic delivery functions. Cell-penetrating peptides (CPPs) are short peptide families that facilitate cellular intake of molecules and vesicles. We previously reported that the modification of cells, and liposomes with CPP-conjugated-lipids, CPPs conjugated with poly (ethylene glycol)-conjugated phospholipids (PEG-lipid), that induce adhesion by CPPs, can be useful for cell-based assays and harvesting liposomes. In this study, we aimed to modulate the exosome surface using Tat peptide (YGRKKRRQRRR)-PEG-lipids to improve intracellular delivery to endothelial cells. We isolated and characterized exosomes from the medium of HEK 293 T cell cultures. Tat conjugated PEG-lipids with different spacer molecular weights and lipid types were incorporated into exosomes using fluorescein isothiocyanate labeling to optimize the number of Tat-PEG-lipids immobilized on the exosome surface. The exosomes modified with Tat-PEG-lipids were incubated with human umbilical vein endothelial cells (HUVECs) to study the interaction. Tat conjugated with 5 kDa PEG and C16 lipids incorporated on the exosome surface were highly detected inside HUVECs by flow cytometry. Fluorescence was negligible in HUVECs for control groups. Thus, Tat-PEG-lipids can be modified on the exosome surface, improving the intracellular delivery of exosomes.
Collapse
Affiliation(s)
- Tianwei Huang
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuya Sato
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Akiko Kuramochi
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Yoshio Ohba
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Masayuki Sano
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Makoto Miyagishi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Hiroaki Tateno
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science & Technology (AIST), Central 5-41, Tsukuba 305-8565, Japan,School of Integrative & Global Majors (SIGMA), Tsukuba Life Science Innovation, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Kazunori Kawasaki
- Material Science RG, Research Institute of Electrochemical Energy, Department of Energy and Environment, National Institute of Advanced Industrial Science and Technology (AIST) 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Takeyuki Uchida
- Material Science RG, Research Institute of Electrochemical Energy, Department of Energy and Environment, National Institute of Advanced Industrial Science and Technology (AIST) 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Kristina N. Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden,Linnaeus Center of Biomaterials Chemistry, Linnaeus University, SE-391 82 Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden
| | - Ung-il Chung
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuji Teramura
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan,Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden,Master's/Doctoral Program in Life Science Innovation (T-LSI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577 Japan,Corresponding author. Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan.
| |
Collapse
|
97
|
Szymoński K, Chmura Ł, Lipiec E, Adamek D. Vibrational spectroscopy – are we close to finding a solution for early pancreatic cancer diagnosis? World J Gastroenterol 2023; 29:96-109. [PMID: 36683712 PMCID: PMC9850953 DOI: 10.3748/wjg.v29.i1.96] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 01/04/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive and lethal neoplasm, ranking seventh in the world for cancer deaths, with an overall 5-year survival rate of below 10%. The knowledge about PC pathogenesis is rapidly expanding. New aspects of tumor biology, including its molecular and morphological heterogeneity, have been reported to explain the complicated “cross-talk” that occurs between the cancer cells and the tumor stroma or the nature of pancreatic ductal adenocarcinoma-associated neural remodeling. Nevertheless, currently, there are no specific and sensitive diagnosis options for PC. Vibrational spectroscopy (VS) shows a promising role in the development of early diagnosis technology. In this review, we summarize recent reports about improvements in spectroscopic methodologies, briefly explain and highlight the drawbacks of each of them, and discuss available solutions. The important aspects of spectroscopic data evaluation with multivariate analysis and a convolutional neural network methodology are depicted. We conclude by presenting a study design for systemic verification of the VS-based methods in the diagnosis of PC.
Collapse
Affiliation(s)
- Krzysztof Szymoński
- Department of Pathomorphology, Jagiellonian University Medical College, Cracow 33-332, Poland
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
| | - Łukasz Chmura
- Department of Pathomorphology, Jagiellonian University Medical College, Cracow 33-332, Poland
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
| | - Ewelina Lipiec
- M. Smoluchowski Institute of Physics, Jagiellonian University, Cracow 30-348, Poland
| | - Dariusz Adamek
- Department of Pathomorphology, University Hospital in Cracow, Cracow 31-501, Poland
- Department of Neuropathology, Jagiellonian University Medical College, Cracow 33-332, Poland
| |
Collapse
|
98
|
Kluszczynska K, Czyz M. Extracellular Vesicles-Based Cell-Cell Communication in Melanoma: New Perspectives in Diagnostics and Therapy. Int J Mol Sci 2023; 24:ijms24020965. [PMID: 36674479 PMCID: PMC9865538 DOI: 10.3390/ijms24020965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of cell-secreted particles that carry cargo of functional biomolecules crucial for cell-to-cell communication with both physiological and pathophysiological consequences. In this review, we focus on evidence demonstrating that the EV-mediated crosstalk between melanoma cells within tumor, between melanoma cells and immune and stromal cells, promotes immune evasion and influences all steps of melanoma development from local progression, pre-metastatic niche formation, to metastatic colonization of distant organs. We also discuss the role of EVs in the development of resistance to immunotherapy and therapy with BRAFV600/MEK inhibitors, and shortly summarize the recent advances on the potential applications of EVs in melanoma diagnostics and therapy.
Collapse
|
99
|
Visan KS, Wu LY, Voss S, Wuethrich A, Möller A. Status quo of Extracellular Vesicle isolation and detection methods for clinical utility. Semin Cancer Biol 2023; 88:157-171. [PMID: 36581020 DOI: 10.1016/j.semcancer.2022.12.008] [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: 09/13/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 12/28/2022]
Abstract
Extracellular vesicles (EVs) are nano-sized particles that hold tremendous potential in the clinical space, as their biomolecular profiles hold a key to non-invasive liquid biopsy for cancer diagnosis and prognosis. EVs are present in most bodily fluids, hence are easily obtainable from patients, advantageous to that of traditional, invasive tissue biopsies and imaging techniques. However, there are certain constraints that hinder clinical use of EVs. The translation of EV biomarkers from "bench-to-bedside" is encumbered by the methods of EV isolation and subsequent biomarker detection currently implemented in laboratories. Although current isolation and detection methods are effective, they lack practicality, with their requirement for high bodily fluid volumes, low equipment availability, slow turnaround times and high costs. The high demand for techniques that overcome these limitations has resulted in significant advancements in nanotechnological devices. These devices are designed to integrate EV isolation and biomarker detection into a one-step method of direct EV detection from bodily fluids. This provides promise for the acceleration of EVs into current clinical standards. This review highlights the importance of EVs as cancer biomarkers, the methodological obstacles currently faced in clinical studies and how novel nanodevices could advance clinical translation.
Collapse
Affiliation(s)
- Kekoolani S Visan
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Li-Ying Wu
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - Sarah Voss
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland 4059, Australia
| | - Alain Wuethrich
- Centre for Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andreas Möller
- Tumour Microenvironment Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; Department of Otorhinolaryngology, Head and Neck Surgery, Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
100
|
Lopez K, Lai SWT, Lopez Gonzalez EDJ, Dávila RG, Shuck SC. Extracellular vesicles: A dive into their role in the tumor microenvironment and cancer progression. Front Cell Dev Biol 2023; 11:1154576. [PMID: 37025182 PMCID: PMC10071009 DOI: 10.3389/fcell.2023.1154576] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Extracellular vesicles (EVs) encompass a diverse set of membrane-derived particles released from cells and are found in numerous biological matrices and the extracellular space. Specific classes of EVs include apoptotic bodies, exosomes, and microvesicles, which vary in their size, origin, membrane protein expression, and interior cargo. EVs provide a mechanism for shuttling cargo between cells, which can influence cell physiology by transporting proteins, DNA, and RNA. EVs are an abundant component of the tumor microenvironment (TME) and are proposed to drive tumor growth and progression by communicating between fibroblasts, macrophages, and tumor cells in the TME. The cargo, source, and type of EV influences the pro- or anti-tumoral role of these molecules. Therefore, robust EV isolation and characterization techniques are required to ensure accurate elucidation of their association with disease. Here, we summarize different EV subclasses, methods for EV isolation and characterization, and a selection of current clinical trials studying EVs. We also review key studies exploring the role and impact of EVs in the TME, including how EVs mediate intercellular communication, drive cancer progression, and remodel the TME.
Collapse
|