1
|
Tuscharoenporn T, Apaijai N, Charoenkwan K, Chattipakorn N, Chattipakorn SC. Emerging roles of exosomes in diagnosis, prognosis, and therapeutic potential in ovarian cancer: a comprehensive review. Cancer Gene Ther 2025:10.1038/s41417-025-00871-2. [PMID: 39843770 DOI: 10.1038/s41417-025-00871-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Ovarian cancer is a leading cause of cancer-related deaths in women, and the development of chemoresistance remains a major challenge during and after its treatment. Exosomes, small extracellular vesicles involved in intercellular communication, have emerged as potential biomarkers and therapeutic targets in ovarian cancer. This review summarizes the current literature on differences in exosomal protein/gene expression between chemosensitive and chemoresistant ovarian cancer, and the effects of exosomal modifications on chemotherapeutic response. Clinical studies have identified alterations in several exosomal components from ovarian cancer tissues and serum samples arising as a consequence of chemosensitivity, which indicates their potential usefulness as potential biomarkers for predicting the development of chemoresistance. Interventional investigations from in vitro and in vivo studies demonstrated that modulation of specific exosomal components can influence ovarian cancer cell phenotypes and individual responses to chemotherapy. Exosomal delivery of chemotherapeutic agents, such as cisplatin, has presented as a potential targeted drug delivery strategy for overcoming chemoresistance in preclinical models. In summary, this review highlights the potential for exosomal proteins and genes to be useful biomarkers for predicting chemotherapy response and being therapeutic targets for overcoming chemoresistance in ovarian cancer. However, future research is still needed to validate these findings and explore the clinical utility of exosomal biomarkers and therapeutics in ovarian cancer management. In addition, understanding the molecular mechanisms underlying exosome-mediated chemoresistance may provide valuable insights for the development of personalized therapeutic strategies, improving outcomes for patients with ovarian cancer.
Collapse
Affiliation(s)
- Thunwipa Tuscharoenporn
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kittipat Charoenkwan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Shao M, Gao Y, Xu X, Chan DW, Du J. Exosomes: Key Factors in Ovarian Cancer Peritoneal Metastasis and Drug Resistance. Biomolecules 2024; 14:1099. [PMID: 39334866 PMCID: PMC11430201 DOI: 10.3390/biom14091099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cancer remains a leading cause of death among gynecological cancers, largely due to its propensity for peritoneal metastasis and the development of drug resistance. This review concentrates on the molecular underpinnings of these two critical challenges. We delve into the role of exosomes, the nano-sized vesicles integral to cellular communication, in orchestrating the complex interactions within the tumor microenvironment that facilitate metastatic spread and thwart therapeutic efforts. Specifically, we explore how exosomes drive peritoneal metastasis by promoting epithelial-mesenchymal transition in peritoneal mesothelial cells, altering the extracellular matrix, and supporting angiogenesis, which collectively enable the dissemination of cancer cells across the peritoneal cavity. Furthermore, we dissect the mechanisms by which exosomes contribute to the emergence of drug resistance, including the sequestration and expulsion of chemotherapeutic agents, the horizontal transfer of drug resistance genes, and the modulation of critical DNA repair and apoptotic pathways. By shedding light on these exosome-mediated processes, we underscore the potential of exosomal pathways as novel therapeutic targets, offering hope for more effective interventions against ovarian cancer's relentless progression.
Collapse
Affiliation(s)
- Ming Shao
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
| | - Yunran Gao
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Xiling Xu
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - David Wai Chan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Juan Du
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
3
|
Skryabin GO, Komelkov AV, Zhordania KI, Bagrov DV, Enikeev AD, Galetsky SA, Beliaeva AA, Kopnin PB, Moiseenko AV, Senkovenko AM, Tchevkina EM. Integrated miRNA Profiling of Extracellular Vesicles from Uterine Aspirates, Malignant Ascites and Primary-Cultured Ascites Cells for Ovarian Cancer Screening. Pharmaceutics 2024; 16:902. [PMID: 39065600 PMCID: PMC11280431 DOI: 10.3390/pharmaceutics16070902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Extracellular vesicles (EVs) are of growing interest in the context of screening for highly informative cancer markers. We have previously shown that uterine aspirate EVs (UA EVs) are a promising source of ovarian cancer (OC) diagnostic markers. In this study, we first conducted an integrative analysis of EV-miRNA profiles from UA, malignant ascitic fluid (AF), and a conditioned medium of cultured ascites cells (ACs). Using three software packages, we identified 79 differentially expressed miRNAs (DE-miRNAs) in UA EVs from OC patients and healthy individuals. To narrow down this panel and select miRNAs most involved in OC pathogenesis, we aligned these molecules with the DE-miRNA sets obtained by comparing the EV-miRNA profiles from OC-related biofluids with the same control. We found that 76% of the DE-miRNAs from the identified panel are similarly altered (differentially co-expressed) in AF EVs, as are 58% in AC EVs. Interestingly, the set of miRNAs differentially co-expressed in AF and AC EVs strongly overlaps (40 out of 44 miRNAs). Finally, the application of more rigorous criteria for DE assessment, combined with the selection of miRNAs that are differentially co-expressed in all biofluids, resulted in the identification of a panel of 29 miRNAs for ovarian cancer screening.
Collapse
Affiliation(s)
- Gleb O. Skryabin
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Andrei V. Komelkov
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Kirill I. Zhordania
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Dmitry V. Bagrov
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia; (D.V.B.); (A.V.M.); (A.M.S.)
| | - Adel D. Enikeev
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Sergey A. Galetsky
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Anastasiia A. Beliaeva
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia; (D.V.B.); (A.V.M.); (A.M.S.)
| | - Pavel B. Kopnin
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| | - Andey V. Moiseenko
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia; (D.V.B.); (A.V.M.); (A.M.S.)
| | - Alexey M. Senkovenko
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow 119991, Russia; (D.V.B.); (A.V.M.); (A.M.S.)
| | - Elena M. Tchevkina
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Highway, Moscow 115522, Russia; (G.O.S.); (A.V.K.); (K.I.Z.); (A.D.E.); (S.A.G.); (A.A.B.); (P.B.K.)
| |
Collapse
|
4
|
Wilczyński J, Paradowska E, Wilczyńska J, Wilczyński M. Prediction of Chemoresistance-How Preclinical Data Could Help to Modify Therapeutic Strategy in High-Grade Serous Ovarian Cancer. Curr Oncol 2023; 31:229-249. [PMID: 38248100 PMCID: PMC10814576 DOI: 10.3390/curroncol31010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is one of the most lethal tumors generally and the most fatal cancer of the female genital tract. The approved standard therapy consists of surgical cytoreduction and platinum/taxane-based chemotherapy, and of targeted therapy in selected patients. The main therapeutic problem is chemoresistance of recurrent and metastatic HGSOC tumors which results in low survival in the group of FIGO III/IV. Therefore, the prediction and monitoring of chemoresistance seems to be of utmost importance for the improvement of HGSOC management. This type of cancer has genetic heterogeneity with several subtypes being characterized by diverse gene signatures and disturbed peculiar epigenetic regulation. HGSOC develops and metastasizes preferentially in the specific intraperitoneal environment composed mainly of fibroblasts, adipocytes, and immune cells. Different HGSOC subtypes could be sensitive to distinct sets of drugs. Moreover, primary, metastatic, and recurrent tumors are characterized by an individual biology, and thus diverse drug responsibility. Without a precise identification of the tumor and its microenvironment, effective treatment seems to be elusive. This paper reviews tumor-derived genomic, mutational, cellular, and epigenetic biomarkers of HGSOC drug resistance, as well as tumor microenvironment-derived biomarkers of chemoresistance, and discusses their possible use in the novel complex approach to ovarian cancer therapy and monitoring.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| | - Justyna Wilczyńska
- Department of Tele-Radiotherapy, Mikolaj Kopernik Provincial Multi-Specialized Oncology and Traumatology Center, 62 Pabianicka Str., 93-513 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| |
Collapse
|
5
|
Mukherjee S, Nag S, Mukerjee N, Maitra S, Muthusamy R, Fuloria NK, Fuloria S, Adhikari MD, Anand K, Thorat N, Subramaniyan V, Gorai S. Unlocking Exosome-Based Theragnostic Signatures: Deciphering Secrets of Ovarian Cancer Metastasis. ACS OMEGA 2023; 8:36614-36627. [PMID: 37841156 PMCID: PMC10568589 DOI: 10.1021/acsomega.3c02837] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/21/2023] [Indexed: 10/17/2023]
Abstract
Ovarian cancer (OC) is a common gynecological cancer worldwide. Unfortunately, the lack of early detection methods translates into a substantial cohort of women grappling with the pressing health crisis. The discovery of extracellular vesicles (EVs) (their major subpopulation exosomes, microvesicles, and apoptotic bodies) has provided new insights into the understanding of cancer. Exosomes, a subpopulation of EVs, play a crucial role in cellular communication and reflect the cellular status under both healthy and pathological conditions. Tumor-derived exosomes (TEXs) dynamically influence ovarian cancer progression by regulating uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and the development of drug and therapeutic resistance. In the field of OC diagnostics, TEXs offer potential biomarkers in various body fluids. On the other hand, exosomes have also shown promising abilities to cure ovarian cancer. In this review, we address the interlink between exosomes and ovarian cancer and explore their theragnostic signature. Finally, we highlight future directions of exosome-based ovarian cancer research.
Collapse
Affiliation(s)
- Sayantanee Mukherjee
- Centre
for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, Kerala, India
| | - Sagnik Nag
- Department
of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Road, Tamil Nadu 632014, India
| | - Nobendu Mukerjee
- Department
of Microbiology, West Bengal State University, West Bengal 700126, Kolkata, India
- Department
of Health Sciences, Novel Global Community
Educational Foundation, New South
Wales, Australia
| | - Swastika Maitra
- Department
of Microbiology, Adamas University, West Bengal 700126, Kolkata, India
| | - Raman Muthusamy
- Department
of Microbiology, Centre for Infectious Diseases, Saveetha Dental College, Chennai, Tamil Nadu 600077, India
| | - Neeraj Kumar Fuloria
- Faculty
of Pharmacy, & Centre of Excellence for Biomaterials Engineering, AIMST University, Semeling, Kedah 08100, Malaysia
| | - Shivkanya Fuloria
- Faculty
of Pharmacy, AIMST University, Semeling, Kedah 08100, Malaysia
| | - Manab Deb Adhikari
- Department
of Biotechnology, University of North Bengal
Raja Rammohunpur, Darjeeling, West Bengal 734013, India
| | - Krishnan Anand
- Department
of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Nanasaheb Thorat
- Limerick
Digital Cancer Research Centre and Department of Physics, Bernal Institute, University of Limerick, Castletroy Co. Limerick, Limerick V94T9PX, Ireland
| | - Vetriselvan Subramaniyan
- Jeffrey
Cheah School of Medicine and Health Sciences, Monash University, Malaysia, Jalan Lagoon Selatan, Bandar
Sunway, 47500 Selangor
Darul Ehsan, Malaysia
- Center
for Transdisciplinary Research, Department of Pharmacology, Saveetha
Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Sukhamoy Gorai
- Rush
University Medical Center, 1620 West Harrison Street, Chicago, Illinois 60612, United States
| |
Collapse
|
6
|
Černe K, Kelhar N, Resnik N, Herzog M, Vodnik L, Veranič P, Kobal B. Characteristics of Extracellular Vesicles from a High-Grade Serous Ovarian Cancer Cell Line Derived from a Platinum-Resistant Patient as a Potential Tool for Aiding the Prediction of Responses to Chemotherapy. Pharmaceuticals (Basel) 2023; 16:907. [PMID: 37375854 DOI: 10.3390/ph16060907] [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: 05/02/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Platinum-resistant high-grade serous ovarian cancer (HGSOC) is invariably a fatal disease. A central goal of ovarian cancer research is therefore to develop new strategies to overcome platinum resistance. Treatment is thus moving towards personalized therapy. However, validated molecular biomarkers that predict patients' risk of developing platinum resistance are still lacking. Extracellular vesicles (EVs) are promising candidate biomarkers. EpCAM-specific EVs are largely unexplored biomarkers for predicting chemoresistance. Using transmission electron microscopy, nanoparticle tracking analysis and flow cytometry, we compared the characteristics of EVs released from a cell line derived from a clinically confirmed cisplatin-resistant patient (OAW28) and EVs released from two cell lines from tumors sensitive to platinum-based chemotherapy (PEO1 and OAW42). We demonstrated that EVs released from the HGSOC cell line of chemoresistant patients exhibited greater size heterogeneity, a larger proportion of medium/large (>200 nm) Evs and a higher number of released EpCAM-positive EVs of different sizes, although the expression of EpCAM was predominant in EVs larger than 400 nm. We also found a strong positive correlation between the concentration of EpCAM-positive EVs and the expression of cellular EpCAM. These results may contribute to the prediction of platinum resistance in the future, although they should first be validated in clinical samples.
Collapse
Affiliation(s)
- Katarina Černe
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Nuša Kelhar
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Nataša Resnik
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Maruša Herzog
- Division of Gynecology and Obstetrics, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
- Department of Gynecology and Obstetrics, Faculty of Medicine, University Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Lana Vodnik
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Peter Veranič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Borut Kobal
- Division of Gynecology and Obstetrics, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
- Department of Gynecology and Obstetrics, Faculty of Medicine, University Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Zheng F, Wang J, Wang D, Yang Q. Clinical Application of Small Extracellular Vesicles in Gynecologic Malignancy Treatments. Cancers (Basel) 2023; 15:cancers15071984. [PMID: 37046644 PMCID: PMC10093031 DOI: 10.3390/cancers15071984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are the key mediators of intercellular communication. They have the potential for clinical use as diagnostic or therapeutic biomarkers and have been explored as vectors for drug delivery. Identification of reliable and noninvasive biomarkers, such as sEVs, is important for early diagnosis and precise treatment of gynecologic diseases to improve patient prognosis. Previous reviews have summarized routine sEVs isolation and identification methods; however, novel and unconventional methods have not been comprehensively described. This review summarizes a convenient method of isolating sEVs from body fluids and liquid biopsy-related sEV markers for early, minimally invasive diagnosis of gynecologic diseases. In addition, the characteristics of sEVs as drug carriers and in precision treatment and drug resistance are introduced, providing a strong foundation for identifying novel and potential therapeutic targets for sEV therapy. We propose potential directions for further research on the applications of sEVs in the diagnosis and treatment of gynecologic diseases.
Collapse
|
8
|
Extracellular Vesicles as Drug Targets and Delivery Vehicles for Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14122822. [PMID: 36559315 PMCID: PMC9788152 DOI: 10.3390/pharmaceutics14122822] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs) are particles that are released from cells into the extracellular space both under pathological and normal conditions. It is now well established that cancer cells secrete more EVs compared to non-cancerous cells and that, captivatingly, several proteins that are involved in EV biogenesis and secretion are upregulated in various tumours. Recent studies have revealed that EVs facilitate the interaction between cancer cells and their microenvironment and play a substantial role in the growth of tumours. As EVs are involved in several aspects of cancer progression including angiogenesis, organotropism, pre-metastatic niche formation, fostering of metastasis, and chemoresistance, inhibiting the release of EVs from cancer and the surrounding tumour microenvironment cells has been proposed as an ideal strategy to treat cancer and associated paraneoplastic syndromes. Lately, EVs have shown immense benefits in preclinical settings as a novel drug delivery vehicle. This review provides a brief overview of the role of EVs in various hallmarks of cancer, focusing on (i) strategies to treat cancer by therapeutically targeting the release of tumour-derived EVs and (ii) EVs as valuable drug delivery vehicles. Furthermore, we also outline the drawbacks of the existing anti-cancer treatments and the future prospective of EV-based therapeutics.
Collapse
|
9
|
Yang Q, Xu J, Gu J, Shi H, Zhang J, Zhang J, Chen Z, Fang X, Zhu T, Zhang X. Extracellular Vesicles in Cancer Drug Resistance: Roles, Mechanisms, and Implications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201609. [PMID: 36253096 PMCID: PMC9731723 DOI: 10.1002/advs.202201609] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 09/10/2022] [Indexed: 06/16/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanosized vesicles that mediate cell-to-cell communication via transporting bioactive molecules and thus are critically involved in various physiological and pathological conditions. EVs contribute to different aspects of cancer progression, such as cancer growth, angiogenesis, metastasis, immune evasion, and drug resistance. EVs induce the resistance of cancer cells to chemotherapy, radiotherapy, targeted therapy, antiangiogenesis therapy, and immunotherapy by transferring specific cargos that affect drug efflux and regulate signaling pathways associated with epithelial-mesenchymal transition, autophagy, metabolism, and cancer stemness. In addition, EVs modulate the reciprocal interaction between cancer cells and noncancer cells in the tumor microenvironment (TME) to develop therapy resistance. EVs are detectable in many biofluids of cancer patients, and thus are regarded as novel biomarkers for monitoring therapy response and predicting prognosis. Moreover, EVs are suggested as promising targets and engineered as nanovehicles to deliver drugs for overcoming drug resistance in cancer therapy. In this review, the biological roles of EVs and their mechanisms of action in cancer drug resistance are summarized. The preclinical studies on using EVs in monitoring and overcoming cancer drug resistance are also discussed.
Collapse
Affiliation(s)
- Qiurong Yang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jing Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory MedicineNantong Tumor HospitalNantongJiangsu226361China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jiayin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jianye Zhang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical PharmacologySchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdong511436China
| | - Zhe‐Sheng Chen
- College of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Xinjian Fang
- Department of OncologyLianyungang Hospital Affiliated to Jiangsu UniversityLianyungangJiangsu222000China
| | - Taofeng Zhu
- Department of Pulmonary and Critical Care MedicineYixing Hospital affiliated to Jiangsu UniversityYixingJiangsu214200China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| |
Collapse
|
10
|
Li J, Li X, Guo Q. Drug Resistance in Cancers: A Free Pass for Bullying. Cells 2022; 11:3383. [PMID: 36359776 PMCID: PMC9654341 DOI: 10.3390/cells11213383] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The cancer burden continues to grow globally, and drug resistance remains a substantial challenge in cancer therapy. It is well established that cancerous cells with clonal dysplasia generate the same carcinogenic lesions. Tumor cells pass on genetic templates to subsequent generations in evolutionary terms and exhibit drug resistance simply by accumulating genetic alterations. However, recent evidence has implied that tumor cells accumulate genetic alterations by progressively adapting. As a result, intratumor heterogeneity (ITH) is generated due to genetically distinct subclonal populations of cells coexisting. The genetic adaptive mechanisms of action of ITH include activating "cellular plasticity", through which tumor cells create a tumor-supportive microenvironment in which they can proliferate and cause increased damage. These highly plastic cells are located in the tumor microenvironment (TME) and undergo extreme changes to resist therapeutic drugs. Accordingly, the underlying mechanisms involved in drug resistance have been re-evaluated. Herein, we will reveal new themes emerging from initial studies of drug resistance and outline the findings regarding drug resistance from the perspective of the TME; the themes include exosomes, metabolic reprogramming, protein glycosylation and autophagy, and the relates studies aim to provide new targets and strategies for reversing drug resistance in cancers.
Collapse
Affiliation(s)
| | | | - Qie Guo
- The Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
11
|
Onuma T, Asare-Werehene M, Yoshida Y, Tsang BK. Exosomal Plasma Gelsolin Is an Immunosuppressive Mediator in the Ovarian Tumor Microenvironment and a Determinant of Chemoresistance. Cells 2022; 11:cells11203305. [PMID: 36291171 PMCID: PMC9600545 DOI: 10.3390/cells11203305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Ovarian Cancer (OVCA) is the most fatal gynecologic cancer and has a 5-year survival rate less than 45%. This is mainly due to late diagnosis and drug resistance. Overexpression of plasma gelsolin (pGSN) is key contributing factor to OVCA chemoresistance and immunosuppression. Gelsolin (GSN) is a multifunctional protein that regulates the activity of actin filaments by cleavage, capping, and nucleation. Generally, it plays an important role in cytoskeletal remodeling. GSN has three isoforms: cytosolic GSN, plasma GSN (pGSN), and gelsolin-3. Exosomes containing pGSN are released and contribute to the progression of OVCA. This review describes how pGSN overexpression inhibits chemotherapy-induced apoptosis and triggers positive feedback loops of pGSN expression. It also describes the mechanisms by which exosomal pGSN promotes apoptosis and dysfunction in tumor-killing immune cells. A discussion on the potential of pGSN as a prognostic, diagnostic, and therapeutic marker is also presented herein.
Collapse
Affiliation(s)
- Toshimichi Onuma
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan
| | - Meshach Asare-Werehene
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan
- Correspondence: (Y.Y.); (B.K.T.)
| | - Benjamin K. Tsang
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Correspondence: (Y.Y.); (B.K.T.)
| |
Collapse
|
12
|
McAlarnen LA, Gupta P, Singh R, Pradeep S, Chaluvally-Raghavan P. Extracellular vesicle contents as non-invasive biomarkers in ovarian malignancies. Mol Ther Oncolytics 2022; 26:347-359. [PMID: 36090475 PMCID: PMC9420349 DOI: 10.1016/j.omto.2022.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer most commonly presents at an advanced stage where survival is approximately 30% compared with >80% if diagnosed and treated before disease spreads. Diagnostic capabilities have progressed from surgical staging via laparotomy to image-guided biopsies and immunohistochemistry staining, along with advances in technology and medicine. Despite improvements in diagnostic capabilities, population-level screening for ovarian cancer is not recommended. Extracellular vesicles (EVs) are 40–150 nm structures formed when the cellular lipid bilayer invaginates. These structures function in cell signaling, immune responses, cancer progression, and establishing the tumor microenvironment. EVs are found in nearly every bodily fluid, including serum, plasma, ascites, urine, and effusion fluid, and contain molecular cargo from their cell of origin. This cargo can be analyzed to yield information about a possible malignancy. In this review we describe how the cargo of EVs has been studied as biomarkers in ovarian cancer. We bring together studies analyzing evidence for various cargos as ovarian cancer biomarkers. Then, we describe the role of EVs in modulation of the tumor microenvironment. This review also summarizes the therapeutic and translational potential of EVs for their optimal utilization as non-invasive biomarkers for novel treatments against cancer.
Collapse
|
13
|
MiR-4284 inhibits sensitivity to paclitaxel in human ovarian carcinoma SKOV3ip1 and HeyA8 cells by targeting DMC1. Anticancer Drugs 2022; 33:701-709. [PMID: 35946537 DOI: 10.1097/cad.0000000000001314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
An increasing number of studies have confirmed that microRNAs (miRNAs) are involved in various biological processes, including tumor growth and drug resistance. MiR-4284 has been proved to be abnormally regulated in several cancers, but the function of miR-4284 in ovarian carcinoma (OC) is unclear. Paclitaxel resistance is a key obstacle in OC treatment. Here, the role of miR-4284 in cell sensitivity to paclitaxel in OC was investigated. Two OC cell lines (SKOV3ip1 and HeyA8) were utilized for the establishment of paclitaxel-resistant cell lines. Reverse transcription-quantitative PCR (RT-qPCR) was applied to analyze the levels of miR-4284 and potential mRNAs in OC cell lines. Western blotting was performed to evaluate the levels of DNA meiotic recombinase 1 (DMC1) protein and cell cycle-associated proteins. Identification of the relationship between miR-4284 and DMC1 was achieved by luciferase reporter assay. CCK-8 and flow cytometry assays were utilized for evaluating the impact of miR-4284 on the malignant characteristics of paclitaxel-resistant OC cells. MiR-4284 was upregulated in paclitaxel-resistant OC cell lines and correlated with an adverse prognosis in OC patients. Depletion of miR-4284 suppressed cell proliferation and cell cycle progression of paclitaxel-resistant OC. MiR-4284 targeted DMC1 which was downregulated in paclitaxel-resistant cells and reversed the inhibitory influence of miR-4284 silencing on the malignant characters of paclitaxel-resistant OC cells. MiR-4284 targets DMC1 to suppress sensitivity to paclitaxel in human OC cells.
Collapse
|
14
|
Fang W, Xia Y. LncRNA HLA-F-AS1 attenuates the ovarian cancer development by targeting miR-21-3p/PEG3 axis. Anticancer Drugs 2022; 33:671-681. [PMID: 35276697 DOI: 10.1097/cad.0000000000001288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dysregulated long noncoding RNA (lncRNA) HLA-F-AS1 is depicted in numerous cancers. However, its function in ovarian cancer has yet to be clarified. LncRNA HLA-F-AS1, miR-21-3p, and PEG3 expressions in ovarian cancer tissues and cells were measured via reverse transcription quantitative PCR. Scratch and CCK8 assays were performed to evaluate the cells' migratory and proliferative abilities, respectively. To assess the expressions of the apoptosis-related proteins Bax and Bcl-2, Western blotting was conducted. Anti-AGO2 RNA immunoprecipitation (RIP) and dual-luciferase reporter assays were executed to study lncRNA HLA-F-AS1's and PEG3 3'UTR's interactions to miR-21-3p. Finally, the tumor growth in vivo was inspected by performing a xenograft experiment. Among the ovarian cancer tissues and cells, the expressions of PEG3 and lncRNA HLA-F-AS1 were depleted while an elevated miR-21-3p expression was observed. HLA-F-AS1's overexpression attenuated ovarian cancer development in vivo and in vitro . MiR-21-3p targeted PEG3 3'UTR while HLA-F-AS1 targeted miR-21-3p. HLA-F-AS1 overexpression mitigated the enhancement brought about by miR-21-3p mimic on ovarian cancer cells' proliferation and migration. Meanwhile, PEG3 overexpression abrogated miR-21-3p mimic's function as an oncogene in the progression of ovarian cancer. Ovarian cancer development is suppressed when lncRNA HLA-F-AS1 targets the miR-21-3p/PEG3 axis. This may possibly be a novel therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Wenhong Fang
- Department of Gynecology and Obstetrics, Wuhan No.6 Hospital, Affiliated Hospital of Jianghan University, Wuhan, Hubei, China
| | | |
Collapse
|
15
|
Liu M, Zhou X, Tang J. Non-Coding RNAs Delivery by Small Extracellular Vesicles and Their Applications in Ovarian Cancer. Front Bioeng Biotechnol 2022; 10:876151. [PMID: 35662846 PMCID: PMC9161355 DOI: 10.3389/fbioe.2022.876151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Ovarian cancer (OC) is the most fatal gynecological malignancy because of its early asymptomatic nature and acquired resistance to chemotherapy. Small extracellular vesicles (sEVs) are a heterogeneous group of biological vesicles with a diameter <200 nm released by cells under physiological or pathological conditions. sEVs-derived non-coding RNAs (ncRNAs) are the essential effectors in the biological environment. sEVs-ncRNAs have critical roles in tumor progression via regulating mRNA expression of target cells to affect cell signaling. In addition, the status of parental cells can be disclosed via analyzing the composition of sEVs-ncRNAs, and their “cargoes” with specific changes can be used as key biomarkers for the diagnosis and prognosis of OC. Accumulating evidence has demonstrated that sEVs-ncRNAs are involved in multiple key processes that mediate the development of metastasis and chemotherapeutic resistance in OC: epithelial–mesenchymal transition; tumorigenicity of mesenchymal stem cells; immune evasion; angiogenesis. The nanomedicine delivery system based on engineering sEVs is expected to be a novel therapeutic strategy for OC. Insights into the biological roles of sEVs-ncRNAs in the invasion, metastasis, immune regulation, and chemoresistance of OC will contribute to discovery of novel biomarkers and molecular targets for early detection and innovative therapy. In this review, we highlight recent advances and applications of sEVs-ncRNAs in OC diagnosis and treatment. We also outline current challenges and knowledge gaps.
Collapse
Affiliation(s)
- Mu Liu
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiaofang Zhou
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jie Tang
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Gynecologic Oncology, Hunan Gynecologic Cancer Research Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Jie Tang,
| |
Collapse
|
16
|
Lee AH, Mejia Peña C, Dawson MR. Comparing the Secretomes of Chemorefractory and Chemoresistant Ovarian Cancer Cell Populations. Cancers (Basel) 2022; 14:1418. [PMID: 35326569 PMCID: PMC8946241 DOI: 10.3390/cancers14061418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) constitutes the majority of all ovarian cancer cases and has staggering rates of both refractory and recurrent disease. While most patients respond to the initial treatment with paclitaxel and platinum-based drugs, up to 25% do not, and of the remaining that do, 75% experience disease recurrence within the subsequent two years. Intrinsic resistance in refractory cases is driven by environmental stressors like tumor hypoxia which alter the tumor microenvironment to promote cancer progression and resistance to anticancer drugs. Recurrent disease describes the acquisition of chemoresistance whereby cancer cells survive the initial exposure to chemotherapy and develop adaptations to enhance their chances of surviving subsequent treatments. Of the environmental stressors cancer cells endure, exposure to hypoxia has been identified as a potent trigger and priming agent for the development of chemoresistance. Both in the presence of the stress of hypoxia or the therapeutic stress of chemotherapy, cancer cells manage to cope and develop adaptations which prime populations to survive in future stress. One adaptation is the modification in the secretome. Chemoresistance is associated with translational reprogramming for increased protein synthesis, ribosome biogenesis, and vesicle trafficking. This leads to increased production of soluble proteins and extracellular vesicles (EVs) involved in autocrine and paracrine signaling processes. Numerous studies have demonstrated that these factors are largely altered between the secretomes of chemosensitive and chemoresistant patients. Such factors include cytokines, growth factors, EVs, and EV-encapsulated microRNAs (miRNAs), which serve to induce invasive molecular, biophysical, and chemoresistant phenotypes in neighboring normal and cancer cells. This review examines the modifications in the secretome of distinct chemoresistant ovarian cancer cell populations and specific secreted factors, which may serve as candidate biomarkers for aggressive and chemoresistant cancers.
Collapse
Affiliation(s)
- Amy H. Lee
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
| | - Carolina Mejia Peña
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| | - Michelle R. Dawson
- Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA;
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA;
| |
Collapse
|
17
|
Ye M, Wang J, Pan S, Zheng L, Wang ZW, Zhu X. Nucleic acids and proteins carried by exosomes of different origins as potential biomarkers for gynecologic cancers. Mol Ther Oncolytics 2022; 24:101-113. [PMID: 35024437 PMCID: PMC8718571 DOI: 10.1016/j.omto.2021.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Miaomiao Ye
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Shuya Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Lihong Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
- Corresponding author Zhi-Wei Wang, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China.
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China
- Corresponding author Xueqiong Zhu, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
18
|
Nanomaterial-based biosensor developing as a route toward in vitro diagnosis of early ovarian cancer. Mater Today Bio 2022; 13:100218. [PMID: 35243293 PMCID: PMC8861407 DOI: 10.1016/j.mtbio.2022.100218] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 12/13/2022] Open
Abstract
The grand challenges of ovarian cancer early diagnosis have led to an alarmingly high mortality rate from ovarian cancer (OC) in the past half century. In vitro diagnosis (IVD) has great potential in the early diagnosis of OC through non-invasive and dynamic analysis of biomarkers. However, common IVDs often fail to provide reliable test results due to lack of sensitivity, specificity, and convenience. In recent years, the discovery of new biomarkers and the progress of nanomaterials can solve the shortcomings of traditional IVD for early OC. These emerging biosensors based on nanomaterials offer great improvements in convenience, speed, selectivity, and sensitivity of IVD. In this review, we firstly systematically summarized the limits of commercial IVD biosensors of OC and the latest discovery of new biomarkers for OC. The representative optimization strategies for six potential ovarian cancer biomarkers are systematically discussed with emphasis on nanomaterial selection and the design of detection principles. Then, various strategies adopted by emerging biosensors based on nanomaterials are also introduced in detail, including optical, electrochemical, microfluidic, and surface plasmon sensors. Finally, current challenges of early OC IVD are proposed, and future research directions on this promising field are also discussed. Failure to diagnose OC early will lead to high mortality. The detection of OC-related biomarkers by IVD method will achieve early diagnosis of OC. The development of nanomaterials-based biosensors is expected to enhance efficiency of detection. Strategies and progress for nanomaterials-based biosensors are systematically reviewed.
Collapse
|
19
|
Comandatore A, Immordino B, Balsano R, Capula M, Garajovà I, Ciccolini J, Giovannetti E, Morelli L. Potential Role of Exosomes in the Chemoresistance to Gemcitabine and Nab-Paclitaxel in Pancreatic Cancer. Diagnostics (Basel) 2022; 12:286. [PMID: 35204377 PMCID: PMC8871170 DOI: 10.3390/diagnostics12020286] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
In recent years, a growing number of studies have evaluated the role of exosomes in pancreatic ductal adenocarcinoma cancer (PDAC) demonstrating their involvement in a multitude of pathways, including the induction of chemoresistance. The aim of this review is to present an overview of the current knowledge on the role of exosomes in the resistance to gemcitabine and nab-paclitaxel, which are two of the most commonly used drugs for the treatment of PDAC patients. Exosomes are vesicular cargos that transport multiple miRNAs, mRNAs and proteins from one cell to another cell and some of these factors can influence specific determinants of gemcitabine activity, such as the nucleoside transporter hENT1, or multidrug resistance proteins involved in the resistance to paclitaxel. Additional mechanisms underlying exosome-mediated resistance include the modulation of apoptotic pathways, cellular metabolism, or the modulation of oncogenic miRNA, such as miR-21 and miR-155. The current status of studies on circulating exosomal miRNA and their possible role as biomarkers are also discussed. Finally, we integrated the preclinical data with emerging clinical evidence, showing how the study of exosomes could help to predict the resistance of individual tumors, and guide the clinicians in the selection of innovative therapeutic strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Benoit Immordino
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Rita Balsano
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Mjriam Capula
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Ingrid Garajovà
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Joseph Ciccolini
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
| |
Collapse
|
20
|
Extracellular vesicles in ovarian cancer chemoresistance, metastasis, and immune evasion. Cell Death Dis 2022; 13:64. [PMID: 35042862 PMCID: PMC8766448 DOI: 10.1038/s41419-022-04510-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/09/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022]
Abstract
Chemoresistance and metastasis are the major challenges for the current ovarian cancer treatment. Understanding the mechanisms of ovarian cancer progression and metastasis is critically important for developing novel therapies. The advances in extracellular vesicles (EVs) research in recent years have attracted extensive attention. EVs contain a variety of proteins, RNAs, DNAs, and metabolites. Accumulating evidence indicates that ovarian cancer cells secrete a large amount of EVs, playing an important role in tumor progression and recurrence. In the microenvironment of ovarian tumor, EVs participate in the information transmission between stromal cells and immune cells, promoting the immune escape of ovarian cancer cells and facilitating cancer metastasis. Here, we review the recent advances of EVs in chemoresistance, mechanisms of metastasis, and immune evasion of ovarian cancer. Furthermore, we also discuss the challenges of EV research and future application of EVs as promising biomarker sources in response to therapy and in therapy-delivery approaches for ovarian cancer patients.
Collapse
|
21
|
Zhang Y, Wei YJ, Zhang YF, Liu HW, Zhang YF. Emerging Functions and Clinical Applications of Exosomal ncRNAs in Ovarian Cancer. Front Oncol 2021; 11:765458. [PMID: 34804970 PMCID: PMC8604153 DOI: 10.3389/fonc.2021.765458] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological malignancies worldwide and has a high mortality rate. Its dismal prognosis is closely related to late diagnosis and drug resistance. Exosomes are a novel means of intercellular communication that are involved in the genesis and development of tumors by delivering a variety of biologically active molecules, including proteins, lipids, and nucleic acids. As an important component, noncoding RNAs (ncRNAs) are selectively enriched in exosomes and participate in the regulation of specific aspects of OC development, such as proliferation, invasion, metastasis, angiogenesis, immune escape, and treatment resistance. Therefore, strategies that specifically target exosomal ncRNAs may be attractive therapeutic options. Exosomes are readily available in almost all types of human biological fluids and are biocompatible, making them promising biomarkers of OC as well as targets for therapeutic applications. In this review, we briefly summarize the biology of exosomes, the function of exosomal ncRNAs in OC development, and their potential clinical applications as biomarkers and therapeutic tools. Ideally, exosomal ncRNAs will become increasingly valuable in the diagnosis and treatment of OC in the near future.
Collapse
Affiliation(s)
- Yu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Jing Wei
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Hao-Wen Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Kalita-de Croft P, Sharma S, Sobrevia L, Salomon C. Extracellular vesicle interactions with the external and internal exposome in mediating carcinogenesis. Mol Aspects Med 2021; 87:101039. [PMID: 34629184 DOI: 10.1016/j.mam.2021.101039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/27/2021] [Indexed: 12/18/2022]
Abstract
The influence of environmental factors on an individual, from conception onwards, is defined as the exposome. It can be categorized into the external exposome, which includes external factors such as air pollution, chemical contaminants, and diet, and the internal exposome, which is unique to an individual, and involves age, physiology, and their genetic profile. The effect of external exposures on the internal exposome, or genetic profile, can be determined through omics analyses. However, this is often compromised due to low sample quantity and cost. Therefore, identification of other factors that can provide an insight into the cellular profile of an individual, provides an exciting avenue, and an emerging field is that of extracellular vesicles (EVs). Recently, our understanding of how cells can communicate with each other has shifted to recognise the role of EVs. EVs are secreted by all living cells, and have been identified in all biological fluids studied so far. They transport bioactive molecules (e.g., proteins, miRNAs, and DNA), and their release can be regulated by the cellular microenvironment. Analysis of EVs in respond to environmental factors might provide novel insights into the role of tumour EVs in carcinogenesis. Not only will EVs give some insight into the tumour cells themselves but they will also provide a better understanding of how cells communicate with one another, contributing to cancer progression. Moreover, characterising the content and functions of tumour-derived EVs has the potential to overcome the current challenges to improve cancer patient outcomes. For example, the identification of EVs targets for therapeutic interventions and tumour EVs biomarkers could facilitate the development of early screening for several cancers. The aim of this review, thus, is to discuss the overall role of EVs in response to the various external and internal signals in cancer. We will specifically highlight the biogenesis, secretion, and content of EVs in response to oncogenic transformation and metabolic regulators in cancer.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Shayna Sharma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, E-41012, Spain; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, 4029, Queensland, Australia; Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen (UMCG), 9713GZ, Groningen, the Netherlands
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, QLD, 4029, Australia; Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
23
|
The Role and Clinical Interest of Extracellular Vesicles in Pregnancy and Ovarian Cancer. Biomedicines 2021; 9:biomedicines9091257. [PMID: 34572444 PMCID: PMC8464910 DOI: 10.3390/biomedicines9091257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer and pregnancy are two states in which the host immune system is exposed to novel antigens. Indeed, both the tumor and placenta must invade tissues, remodel vasculature to establish a robust blood supply, and evade detection by the immune system. Interestingly, tumor and placenta tissue use similar mechanisms to induce these necessary changes. One mediator is emerging as a key player in invasion, vascular remodeling, and immune evasion: extracellular vesicles (EVs). Many studies have identified EVs as a key mediator of cell-to-cell communication. Specifically, the cargo carried by EVs, which includes proteins, nucleic acids, and lipids, can interact with cells to induce changes in the target cell ranging from gene expression to migration and metabolism. EVs can promote cell division and tissue invasion, immunosuppression, and angiogenesis which are essential for both cancer and pregnancy. In this review, we examine the role of EVs in ovarian cancer metastasis, chemoresistance, and immune modulation. We then focus on the role of EVs in pregnancy with special attention on the vascular remodeling and regulation of the maternal immune system. Lastly, we discuss the clinical utility of EVs as markers and therapeutics for ovarian cancer and pre-eclampsia.
Collapse
|
24
|
Campos A, Sharma S, Obermair A, Salomon C. Extracellular Vesicle-Associated miRNAs and Chemoresistance: A Systematic Review. Cancers (Basel) 2021; 13:cancers13184608. [PMID: 34572835 PMCID: PMC8472653 DOI: 10.3390/cancers13184608] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary There is an urgent need for a non-invasive, specific biomarker to identify patients at risk of chemoresistance, which it is the ability of cancer cells to escape the effect of chemotherapy drugs. Extracellular vesicles contain an abundance of miRNAs that demonstrate expression across a range of cancers including breast cancer, renal cell carcinoma, lung cancer, multiple myeloma, and lymphoma. Interestingly, miRNAs encapsulated within extracellular vesicles (EVs) including exosomes display an association with chemoresistance. Here, we performed a systematic revision to evaluate the association between miRNAs within EVs and chemotherapy resistance. The summarized graphical abstract indicates that several exosome-derived miRNAs involved in chemotherapy resistance can be found among different types of cancers, such as colorectal, ovarian, breast, and lung cancer, and lymphoma. Abstract Cancer is a leading public health issue globally, and diagnosis is often associated with poor outcomes and reduced patient survival. One of the major contributors to the fatality resultant of cancer is the development of resistance to chemotherapy, known as chemoresistance. Furthermore, there are limitations in our ability to identify patients that will respond to therapy, versus patients that will develop relapse, and display limited or no response to treatment. This often leads to patients being subjected to multiple futile treatment cycles, and results in a reduction in their quality of life. Therefore, there is an urgent clinical need to develop tools to identify patients at risk of chemoresistance, and recent literature has suggested that small extracellular vesicles, known as exosomes, may be a vital source of information. Extracellular vesicles (EV) are membrane bound vesicles, involved in cell-cell communication, through the transfer of their cargo, which includes proteins, lipids, and miRNAs. A defined exploration strategy was performed in this systematic review in order to provide a compilation of key EV miRNAs which may be predictive of chemoresistance. We searched the PubMed, Science Direct, and Scopus databases using the following keywords: Extracellular vesicles OR exosomes OR EVs AND miRNA AND Chemotherapy OR Chemoresistance OR Cancer Recurrence from 2010 to 2020. We found 31 articles that reported key EV-associated miRNAs involved in cancer recurrence related to chemoresistance. Interestingly, multiple studies of the same tumor type identified different microRNAs, and few studies identified the same ones. Specifically, miR-21, miR-222, and miR-155 displayed roles in response to chemotherapy, and were found to be common in colorectal cancer, ovarian cancer, breast cancer, and diffuse large B cell lymphoma patients (DLBCL). miR-21 and miR-222 were found to favour the development of chemoresistance, whereas miR-155 exhibited a contrasting role, depending on the type of primary tumor. Whilst high levels of miR-155 were found to correlate with chemotherapy resistance in DLBCL, it was found to be predictive of an effective response towards chemotherapy in breast cancer. Thus, further research regarding the roles of these miRNAs would be beneficial in terms of designing novel tools to counteract the progression of cancer in a not-to-distant future.
Collapse
Affiliation(s)
- America Campos
- Exosome Biology Laboratory, Center for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, QLD 4029, Australia; (A.C.); (S.S.)
| | - Shayna Sharma
- Exosome Biology Laboratory, Center for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, QLD 4029, Australia; (A.C.); (S.S.)
| | - Andreas Obermair
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, Centre for Clinical Research, Building 71/918, Royal Brisbane and Women’s Hospital, Herston, QLD 4029, Australia;
| | - Carlos Salomon
- Exosome Biology Laboratory, Center for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, QLD 4029, Australia; (A.C.); (S.S.)
- Departamento de Investigación, Postgrado y Educación Contínua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago 8370007, Chile
- Correspondence: ; Tel.: +61-7-33465500; Fax: +61-7-3346-5509
| |
Collapse
|
25
|
Croft PKD, Sharma S, Godbole N, Rice GE, Salomon C. Ovarian-Cancer-Associated Extracellular Vesicles: Microenvironmental Regulation and Potential Clinical Applications. Cells 2021; 10:cells10092272. [PMID: 34571921 PMCID: PMC8471580 DOI: 10.3390/cells10092272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/08/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is one of the most diagnosed gynecological cancers in women. Due to the lack of effective early stage screening, women are more often diagnosed at an advanced stage; therefore, it is associated with poor patient outcomes. There are a lack of tools to identify patients at the highest risk of developing this cancer. Moreover, early detection strategies, therapeutic approaches, and real-time monitoring of responses to treatment to improve survival and quality of life are also inadequate. Tumor development and progression are dependent upon cell-to-cell communication, allowing cancer cells to re-program cells not only within the surrounding tumor microenvironment, but also at distant sites. Recent studies established that extracellular vesicles (EVs) mediate bi-directional communication between normal and cancerous cells. EVs are highly stable membrane vesicles that are released from a wide range of cells, including healthy and cancer cells. They contain tissue-specific signaling molecules (e.g., proteins and miRNA) and, once released, regulate target cell phenotypes, inducing a pro-tumorigenic and immunosuppressive phenotype to contribute to tumor growth and metastasis as well as proximal and distal cell function. Thus, EVs are a “fingerprint” of their cell of origin and reflect the metabolic status. Additionally, via the capacity to evade the immune system and remain stable over long periods in circulation, EVs can be potent therapeutic agents. This review examines the potential role of EVs in the different aspects of the tumor microenvironment in OC, as well as their application in diagnosis, delivery of therapeutic agents, and disease monitoring.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia
| | - Shayna Sharma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Nihar Godbole
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Gregory E. Rice
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4029, Australia; (P.K.-d.C); (S.S); (N.G); (G.E.R)
- Correspondence: ; Tel.: +61-7-3346-5500; Fax: +61-7-3346-5509
| |
Collapse
|
26
|
Hu C, Zhang L, Yang Z, Song Z, Zhang Q, He Y. Graphene oxide-based qRT-PCR assay enables the sensitive and specific detection of miRNAs for the screening of ovarian cancer. Anal Chim Acta 2021; 1174:338715. [PMID: 34247740 DOI: 10.1016/j.aca.2021.338715] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/01/2022]
Abstract
Circulating microRNAs (miRNAs) have the potential to become reliable and noninvasive biomarkers for ovarian cancer (OC) diagnosis; however, the conventional miRNAs detection techniques exhibit enduring limitations of low sensitivity and specificity. Graphene oxide (GO), a novel nanomaterial, is at the forefront of material design for extensive biomedical applications. Owing to the excellent water affinity and single-stranded DNA (ssDNA) adsorption characteristics of GO, we designed and developed a GO-based qRT-PCR assay for the detection of miRNAs associated with OC. In the GO-based qRT-PCR system, GO could significantly improve the sensitivity and specificity of the qRT-PCR assay by noncovalently interacting with primers and ssDNA and reducing the occurrence of non-specific amplification. Moreover, the detection of miRNAs associated with OC confirmed that GO-based qRT-PCR assay could differentiate benign ovarian tumors from OC (sensitivity, 0.91; specificity, 1.00). Collectively, these findings provide robust evidence that GO-based qRT-PCR assay can be effectively used as a promising method to detect miRNAs for the screening of OC patients.
Collapse
Affiliation(s)
- Chenyan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Linlin Zhang
- Department of Gynecology, People's Hospital of Mianzhu City, Deyang, Sichuan, 618200, China
| | - Zhongzhu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Zhen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Qin Zhang
- Department of Gynecology, People's Hospital of Mianzhu City, Deyang, Sichuan, 618200, China
| | - Yang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
27
|
Storti G, Scioli MG, Kim BS, Terriaca S, Fiorelli E, Orlandi A, Cervelli V. Mesenchymal Stem Cells in Adipose Tissue and Extracellular Vesicles in Ovarian Cancer Patients: A Bridge toward Metastatic Diffusion or a New Therapeutic Opportunity? Cells 2021; 10:cells10082117. [PMID: 34440886 PMCID: PMC8392703 DOI: 10.3390/cells10082117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the deadliest malignancies among women. Approximately 75% of the patients with ovarian cancer are diagnosed with advanced disease that already has metastasis, particularly to the omentum. The omentum constitutes the ideal soil for ovarian cancer metastasis due to a complex intraperitoneal milieu that favors and supports the whole metastatic process. Adipose-derived stem/stromal cells (ADSCs) are part of this microenvironment and foster tumor progression via sustained paracrine secretion, including extracellular vesicles (EVs). Nonetheless, the preferential relationship between ADSCs, ADSC-derived EVs, and ovarian cancer cells could be exploited to use ADSCs and EVs as a vehicle for anti-cancer therapies. This review will analyze the strict relations between tumor progression, metastatic disease, and adipose tissue with its staminal components. In addition, we will describe the crosstalk and biologic relationship between ADSCs and tumor cells, the role of EVs in intercellular communication, the establishment of drug resistance, metastatic capacity, and ovarian cancer progression. We will analyze the new therapeutic opportunities in treating ovarian cancer offered by ADSCs and EVs as a vehicle for therapeutic molecules to target precisely tumor cells and limit the systemic adverse effects. Finally, we will discuss the limitations of these therapeutic approaches.
Collapse
Affiliation(s)
- Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy;
- Correspondence: ; Tel.: +39-06-23188514; Fax: +39-06-23188466
| | - Maria Giovanna Scioli
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Sonia Terriaca
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Elena Fiorelli
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Augusto Orlandi
- Department of Biomedicine and Prevention, Anatomic Pathology Institute, Tor Vergata University of Rome, 00133 Rome, Italy; (M.G.S.); (S.T.); (E.F.); (A.O.)
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy;
| |
Collapse
|
28
|
Umer M, Aziz NB, Mahmudunnabi RG, Shim YB, Salomon C, Shiddiky MJA. A novel DNA binding protein-based platform for electrochemical detection of miRNA. Analyst 2021; 146:5496-5501. [PMID: 34346412 DOI: 10.1039/d1an00935d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We present a novel amplification-free sandwich type platform assay for electrochemical detection of miRNA. The assay is based on T4 DNA polymerase mediated synthesis of the p53 binding DNA sequence at the 3' end of target miRNA. The resulting miRNA-DNA chimera is detected via an electrochemical sandwich hybridization assay where HRP-labelled p53 binds to its recognition sequence and an amperometric signal is generated by hydroquinone-mediated enzymatic reduction of H2O2. The limit of detection of our assay was estimated to be 22 fM with a linear dynamic range of 100 fM-1 nM. This new platform method of detecting miRNA shows superior performance to conventional electrochemical miRNA biosensors and has the potential for amplification-free analysis of miRNA with high specificity and sensitivity.
Collapse
Affiliation(s)
- Muhammad Umer
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan, QLD 4111, Australia.
| | - Nahian B Aziz
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, Australia
| | - Rabbee G Mahmudunnabi
- Department of Chemistry and Institute of BioPhysio Sensor Technology, Pusan National University, Busan 46241, South Korea
| | - Yoon-Bo Shim
- Department of Chemistry and Institute of BioPhysio Sensor Technology, Pusan National University, Busan 46241, South Korea
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, Australia
| | - Muhammad J A Shiddiky
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan, QLD 4111, Australia. and School of Environment and Science, Griffith University, QLD 4111, Australia
| |
Collapse
|
29
|
Role of Extracellular Vesicles in Compromising Cellular Resilience to Environmental Stressors. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9912281. [PMID: 34337063 PMCID: PMC8321721 DOI: 10.1155/2021/9912281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs), like exosomes, are nanosized membrane-enveloped vesicles containing different bioactive cargo, such as proteins, lipids, mRNA, miRNA, and other small regulatory RNAs. Cell-derived EVs, including EVs originating from stem cells, may capture components from damaged cells or cells impacted by therapeutic treatments. Interestingly, EVs derived from stem cells can be preconditioned to produce and secrete EVs with different therapeutic properties, particularly with respect to heat-shock proteins and other molecular cargo contents. This behavior is consistent with stem cells that also respond differently to various microenvironments. Heat-shock proteins play roles in cellular protection and mediate cellular resistance to radiotherapy, chemotherapy, and heat shock. This review highlights the possible roles EVs play in mediating cellular plasticity and survival when exposed to different physical and chemical stressors, with a special focus on the respiratory distress due to the air pollution.
Collapse
|
30
|
Cao J, Zhang Y, Mu J, Yang D, Gu X, Zhang J. Exosomal miR-21-5p contributes to ovarian cancer progression by regulating CDK6. Hum Cell 2021; 34:1185-1196. [PMID: 33813728 DOI: 10.1007/s13577-021-00522-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/17/2021] [Indexed: 12/23/2022]
Abstract
Ovarian cancer is a predominant gynecologic malignancy and correlated with high mortality and severe morbidity. Exosomal microRNAs (miRNAs) play crucial roles in various processes during the progression of ovarian cancer, such as cell proliferation, apoptosis, and invasion. However, the function of exosomal miR-21-5p in ovarian cancer is still unknown. Here, we found that miR-21-5p was upregulated in ovarian cancer tissues, plasma exosomes of ovarian cancer patients, and exosomes from ovarian cancer cells. MiR-21-5p was incorporated in the exosomes from the ovarian cancer cells. In addition, 5-ethynyl-2'-deoxyuridine (Edu), a marker of cancer cell proliferation, was enhanced by miR-21-5p mimic while reduced by miR-21-5p inhibitor in ovarian cancer cells. MiR-21-5p mimic could increase, but miR-21-5p inhibitor could decrease the migration and invasion of cancer cells. Ovarian cancer cell apoptosis was induced by miR-21-5p inhibitor. Moreover, miR-21-5p inhibitor could up-regulate the expression of pro-apoptotic cleaved caspase3 and Bax while downregulate the expression of anti-apoptotic Bcl2 in the cells. Exosomal miR-21-5p inhibited the expression of cyclin-dependent kinase 6 (CDK6) by targeting its 3'-untranslated region (3'-UTR) at both the mRNA and protein levels. Tumorigenicity analysis in nude mice revealed that exosomal miR-21-5p could increase tumor volume, size, and weight of ovarian cancer in vivo. Besides, miR-21-5p targeted CDK6 in tumor tissues of nude mice. In conclusion, exosomal miR-21-5p contributes to the progression of ovarian cancer by regulating CDK6. Our findings will provide novel insights into the mechanism of exosomal miR-21-5p in the development of ovarian cancer. Exosomal miR-21-5p may serve as a potential target for the therapy of ovarian cancer.
Collapse
Affiliation(s)
- Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China
| | - Yuan Zhang
- Department of Gynecology Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, Anhui Province, 233000, People's Republic of China
| | - Juan Mu
- Department of Nutrition, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, People's Republic of China
| | - Dazhen Yang
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China
| | - Xiaoyan Gu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, No. 123 Tianfei Lane, Mochou Road, Nanjing City, Jiangsu Province, 210004, People's Republic of China.
| | - Jing Zhang
- Department of Gynecology Oncology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, Anhui Province, 233000, People's Republic of China.
| |
Collapse
|
31
|
Weinman MA, Ramsey SA, Leeper HJ, Brady JV, Schlueter A, Stanisheuski S, Maier CS, Miller T, Ruby CE, Bracha S. Exosomal proteomic signatures correlate with drug resistance and carboplatin treatment outcome in a spontaneous model of canine osteosarcoma. Cancer Cell Int 2021; 21:245. [PMID: 33933069 PMCID: PMC8088716 DOI: 10.1186/s12935-021-01943-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma patients often experience poor outcomes despite chemotherapy treatment, likely due in part to various mechanisms of tumor cell innate and/or acquired drug resistance. Exosomes, microvesicles secreted by cells, have been shown to play a role in drug resistance, but a comprehensive protein signature relating to osteosarcoma carboplatin resistance has not been fully characterized. METHODS In this study, cell lysates and exosomes from two derivatives (HMPOS-2.5R and HMPOS-10R) of the HMPOS osteosarcoma cell line generated by repeated carboplatin treatment and recovery, were characterized proteomically by mass spectrometry. Protein cargos of circulating serum exosomes from dogs with naturally occurring osteosarcoma, were also assessed by mass spectrometry, to identify biomarkers that discriminate between good and poor responders to carboplatin therapy. RESULTS Both cell lysates and exosomes exhibited distinct protein signatures related to drug resistance. Furthermore, exosomes from the resistant HMPOS-2.5R cell line were found to transfer drug resistance to drug-sensitive HMPOS cells. The comparison of serum exosomes from dogs with a favorable disease-free interval [DFI] of > 300 days, and dogs with < 100 days DFI revealed a proteomic signature that could discriminate between the two cohorts with high accuracy. Furthermore, when the patient's exosomes were compared to exosomes isolated from carboplatin resistant cell lines, several putative biomarkers were found to be shared. CONCLUSIONS The findings of this study highlight the significance of exosomes in the potential transfer of drug resistance, and the discovery of novel biomarkers for the development of liquid biopsies to better guide personalized chemotherapy treatment.
Collapse
|
32
|
Javadi M, Rad JS, Farashah MSG, Roshangar L. An Insight on the Role of Altered Function and Expression of Exosomes and MicroRNAs in Female Reproductive Diseases. Reprod Sci 2021; 29:1395-1407. [PMID: 33825167 DOI: 10.1007/s43032-021-00556-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Exosomes are small bilayer-lipid membrane vesicles secreted by living cells that are able to transfer regulatory molecules and genetic information from one cell to another. These vesicles are enriched with several nucleic acids including mRNAs, microRNAs (miRNAs), other non-coding RNAs, as well as proteins and lipids. Alterations in the exosomal content and functions are observed in numerous reproductive diseases in both animals and human cases. MicroRNAs, a class of small endogenous RNA molecules, can negatively regulate gene expression at the post-transcription level. Aberrant microRNA expression has been reported in multiple human reproductive diseases such as polycystic ovary syndrome, preeclampsia, uterine leiomyomata, ovarian cancer, endometriosis, and Asherman's syndrome. This study focuses to review recent research on alterations of microRNA expression and the role of exosomes in female reproductive diseases. It has been demonstrated that exosomes may be a potential therapeutic approach in various female reproductive diseases. In addition, changes in expression of microRNAs act as molecular biomarkers for diagnosis of several reproductive diseases in women, and regulation of their expression can potentially reduce infertility.
Collapse
Affiliation(s)
- Maryam Javadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Gholami Farashah
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Anatomical Sciences, Faculty of medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Bai Y, Guo J, Liu Z, Li Y, Jin S, Wang T. The Role of Exosomes in the Female Reproductive System and Breast Cancers. Onco Targets Ther 2020; 13:12567-12586. [PMID: 33324075 PMCID: PMC7733408 DOI: 10.2147/ott.s281909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Exosomes are nanoscale extracellular vesicles released by nearly all cell types. Exosomes were originally considered as waste receptacles for discarding unwanted cellular products; however, these organelles are now considered to be important for cell communication by delivering biologically active molecules such as proteins, DNA, non-coding RNA and mRNA. Studies have revealed that exosomes are closely related to several diseases, especially cancers. Exosomes are indispensable for the emergence and progression of tumor. Here, we review the status of research on exosomes in the female reproductive system cancers and breast cancer, focusing on their biological roles in chemical resistance and immune responses, as well as their underlying applications in drug delivery and nanotherapy and as biological markers for tumor diagnosis.
Collapse
Affiliation(s)
- Yuqi Bai
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Jie Guo
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhongshan Liu
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Yunfeng Li
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Shunzi Jin
- NHC Key Laboratory of Radiobiology, Jilin University, Changchun, People's Republic of China
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
34
|
Ke R, Lv L, Zhang S, Zhang F, Jiang Y. Functional mechanism and clinical implications of MicroRNA-423 in human cancers. Cancer Med 2020; 9:9036-9051. [PMID: 33174687 PMCID: PMC7724490 DOI: 10.1002/cam4.3557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 08/16/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs play a vital role in the regulatory mechanisms of tumorigenesis. Current research indicates that microRNA-423 (miR-423) is abnormally expressed in various human tumors and participates in multiple signaling pathways of cancer progression. In most studies, miR-423 was confirmed as oncomiR, while a few contradictory reports considered miR-423 as an anticancer miRNA. The paradoxical role in cancer may hinder the application of miR-423 as a diagnostic and therapeutic target. Simultaneously, the interaction mechanism between miR-423 and lncRNA also needs attention. In this review, we have summarized the dual role of aberrant miR-423 expression and its mechanisms in tumorigenesis, and the therapeutic potential of miR-423 in human tumors.
Collapse
Affiliation(s)
- RuiSheng Ke
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China.,The Fuzong Clinical Medical College of Fujian Medical University, Fuzhou City, China
| | - LiZhi Lv
- The Fuzong Clinical Medical College of Fujian Medical University, Fuzhou City, China.,Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistics Team, Fuzhou City, China
| | - SiYu Zhang
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - FuXing Zhang
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yi Jiang
- The Fuzong Clinical Medical College of Fujian Medical University, Fuzhou City, China.,Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistics Team, Fuzhou City, China
| |
Collapse
|
35
|
Effects of miR-373 Inhibition on Glioblastoma Growth by Reducing Limk1 In Vitro. J Immunol Res 2020; 2020:7671502. [PMID: 33062725 PMCID: PMC7539108 DOI: 10.1155/2020/7671502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor with shorter median overall survival time. It is urgent to find novel methods to enhance the therapeutic efficiency clinically. miR-373 is related to the biological development process of cancers, but there are no reports whether modulation on miR-373 could affect GBM development or modify the efficiency of chemo- or radiotherapy yet. Our current study found that the higher level of miR-373 was observed in U-251 cells. Inhibition on miR-373 could reduce the U-251 cell number by 65% and PCNA expression obviously. In addition, inhibition on miR-373 sensitized U-251 cells to chemo- or radiotherapy. The cell cycle of U-251 cells could be modulated by miR-373 knockdown, which could enhance the p21 expression and reduce the cdc2 level. Anti-miR-373 could increase the Bax/Bcl-2 ratio of U-251 cells and induce cell apoptosis significantly. These above effects of miR-373 could be reversed by Limk1 overexpression. Thus, our experimental data confirmed the fact that miR-373 could be a new therapeutic target to enhance the efficiency of chemo- or radiotherapy for clinical GBM patients.
Collapse
|