1
|
Wang Y, Ma Q, Wang T, Xing J, Li Q, Wang D, Wang G. The involvement and application potential of exosomes in breast cancer immunotherapy. Front Immunol 2024; 15:1384946. [PMID: 38835784 PMCID: PMC11148227 DOI: 10.3389/fimmu.2024.1384946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
Breast cancer has a high incidence and a heightened propensity for metastasis. The absence of precise targets for effective intervention makes it imperative to devise enhanced treatment strategies. Exosomes, characterized by a lipid bilayer and ranging in size from 30 to 150 nm, can be actively released by various cells, including those in tumors. Exosomes derived from distinct subsets of immune cells have been shown to modulate the immune microenvironment within tumors and influence breast cancer progression. In addition, tumor-derived exosomes have been shown to contribute to breast cancer development and progression and may become a new target for breast cancer immunotherapy. Tumor immunotherapy has become an option for managing tumors, and exosomes have become therapeutic vectors that can be used for various pathological conditions. Edited exosomes can be used as nanoscale drug delivery systems for breast cancer therapy, contributing to the remodeling of immunosuppressive tumor microenvironments and influencing the efficacy of immunotherapy. This review discusses the regulatory role of exosomes from different cells in breast cancer and the latest applications of exosomes as nanoscale drug delivery systems and immunotherapeutic agents in breast cancer, showing the development prospects of exosomes in the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Yun Wang
- Department of Thoracic Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Qiji Ma
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Tielin Wang
- College of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Jie Xing
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Gang Wang
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Dzhugashvili E, Tamkovich S. Exosomal Cargo in Ovarian Cancer Dissemination. Curr Issues Mol Biol 2023; 45:9851-9867. [PMID: 38132461 PMCID: PMC10742327 DOI: 10.3390/cimb45120615] [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: 10/09/2023] [Revised: 11/22/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Ovarian cancer (OC) has the highest mortality rate among all gynecologic cancers and is characterized by early peritoneal spread. The growth and development of OC are associated with the formation of ascitic fluid, creating a unique tumor microenvironment. Understanding the mechanisms of tumor progression is crucial in identifying new diagnostic biomarkers and developing novel therapeutic strategies. Exosomes, lipid bilayer vesicles measuring 30-150 nm in size, are known to establish a crucial link between malignant cells and their microenvironment. Additionally, the confirmed involvement of exosomes in carcinogenesis enables them to mediate the invasion, migration, metastasis, and angiogenesis of tumor cells. Functionally active non-coding RNAs (such as microRNAs, long non-coding RNAs, circRNAs), proteins, and lipid rafts transported within exosomes can activate numerous signaling pathways and modify gene expression. This review aims to expand our understanding of the role of exosomes and their contents in OC carcinogenesis processes such as epithelial-mesenchymal transition (EMT), angiogenesis, vasculogenic mimicry, tumor cell proliferation, and peritoneal spread. It also discusses the potential for utilizing exosomal cargo to develop novel "liquid biopsy" biomarkers for early OC diagnosis.
Collapse
Affiliation(s)
- Ekaterina Dzhugashvili
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Svetlana Tamkovich
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
3
|
Lee Y, Ni J, Wasinger VC, Graham P, Li Y. Comparison Study of Small Extracellular Vesicle Isolation Methods for Profiling Protein Biomarkers in Breast Cancer Liquid Biopsies. Int J Mol Sci 2023; 24:15462. [PMID: 37895140 PMCID: PMC10607056 DOI: 10.3390/ijms242015462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are an important intercellular communicator, participating in all stages of cancer metastasis, immunity, and therapeutic resistance. Therefore, protein cargoes within sEVs are considered as a superior source for breast cancer (BC) biomarker discovery. Our study aimed to optimise the approach for sEV isolation and sEV proteomic analysis to identify potential sEV protein biomarkers for BC diagnosis. sEVs derived from BC cell lines, BC patients' plasma, and non-cancer controls were isolated using ultracentrifugation (UC), a Total Exosome Isolation kit (TEI), and a combined approach named UCT. In BC cell lines, the UC isolates showed a higher sEV purity and marker expression, as well as a higher number of sEV proteins. In BC plasma samples, the UCT isolates showed the highest proportion of sEV-related proteins and the lowest percentage of lipoprotein-related proteins. Our data suggest that the assessment of both the quantity and quality of sEV isolation methods is important in selecting the optimal approach for the specific sEV research purpose, depending on the sample types and downstream analysis.
Collapse
Affiliation(s)
- Yujin Lee
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Jie Ni
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Valerie C. Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Kensington, NSW 2052, Australia;
| | - Peter Graham
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Yong Li
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
4
|
Muñoz JP, Pérez-Moreno P, Pérez Y, Calaf GM. The Role of MicroRNAs in Breast Cancer and the Challenges of Their Clinical Application. Diagnostics (Basel) 2023; 13:3072. [PMID: 37835815 PMCID: PMC10572677 DOI: 10.3390/diagnostics13193072] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a subclass of non-coding RNAs that exert substantial influence on gene-expression regulation. Their tightly controlled expression plays a pivotal role in various cellular processes, while their dysregulation has been implicated in numerous pathological conditions, including cancer. Among cancers affecting women, breast cancer (BC) is the most prevalent malignant tumor. Extensive investigations have demonstrated distinct expression patterns of miRNAs in normal and malignant breast cells. Consequently, these findings have prompted research efforts towards leveraging miRNAs as diagnostic tools and the development of therapeutic strategies. The aim of this review is to describe the role of miRNAs in BC. We discuss the identification of oncogenic, tumor suppressor and metastatic miRNAs among BC cells, and their impact on tumor progression. We describe the potential of miRNAs as diagnostic and prognostic biomarkers for BC, as well as their role as promising therapeutic targets. Finally, we evaluate the current use of artificial intelligence tools for miRNA analysis and the challenges faced by these new biomedical approaches in its clinical application. The insights presented in this review underscore the promising prospects of utilizing miRNAs as innovative diagnostic, prognostic, and therapeutic tools for the management of BC.
Collapse
Affiliation(s)
- Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| | - Pablo Pérez-Moreno
- Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7780272, Chile
| | - Yasmín Pérez
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| |
Collapse
|
5
|
Murillo Carrasco AG, Otake AH, Macedo-da-Silva J, Feijoli Santiago V, Palmisano G, Andrade LNDS, Chammas R. Deciphering the Functional Status of Breast Cancers through the Analysis of Their Extracellular Vesicles. Int J Mol Sci 2023; 24:13022. [PMID: 37629204 PMCID: PMC10455604 DOI: 10.3390/ijms241613022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) accounts for the highest incidence of tumor-related mortality among women worldwide, justifying the growing search for molecular tools for the early diagnosis and follow-up of BC patients under treatment. Circulating extracellular vesicles (EVs) are membranous nanocompartments produced by all human cells, including tumor cells. Since minimally invasive methods collect EVs, which represent reservoirs of signals for cell communication, these particles have attracted the interest of many researchers aiming to improve BC screening and treatment. Here, we analyzed the cargoes of BC-derived EVs, both proteins and nucleic acids, which yielded a comprehensive list of potential markers divided into four distinct categories, namely, (i) modulation of aggressiveness and growth; (ii) preparation of the pre-metastatic niche; (iii) epithelial-to-mesenchymal transition; and (iv) drug resistance phenotype, further classified according to their specificity and sensitivity as vesicular BC biomarkers. We discuss the therapeutic potential of and barriers to the clinical implementation of EV-based tests, including the heterogeneity of EVs and the available technologies for analyzing their content, to present a consistent, reproducible, and affordable set of markers for further evaluation.
Collapse
Affiliation(s)
- Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Andreia Hanada Otake
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Janaina Macedo-da-Silva
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
| | - Veronica Feijoli Santiago
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
| | - Giuseppe Palmisano
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (J.M.-d.-S.); (V.F.S.); (G.P.)
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (A.H.O.); (L.N.d.S.A.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
6
|
Timofeeva AM, Paramonik AP, Sedykh SS, Nevinsky GA. Milk Exosomes: Next-Generation Agents for Delivery of Anticancer Drugs and Therapeutic Nucleic Acids. Int J Mol Sci 2023; 24:10194. [PMID: 37373342 DOI: 10.3390/ijms241210194] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Exosomes are nanovesicles 40-120 nm in diameter secreted by almost all cell types and providing humoral intercellular interactions. Given the natural origin and high biocompatibility, the potential for loading various anticancer molecules and therapeutic nucleic acids inside, and the surface modification possibility for targeted delivery, exosomes are considered to be a promising means of delivery to cell cultures and experimental animal organisms. Milk is a unique natural source of exosomes available in semi-preparative and preparative quantities. Milk exosomes are highly resistant to the harsh conditions of the gastrointestinal tract. In vitro studies have demonstrated that milk exosomes have an affinity to epithelial cells, are digested by cells by endocytosis mechanism, and can be used for oral delivery. With milk exosome membranes containing hydrophilic and hydrophobic components, exosomes can be loaded with hydrophilic and lipophilic drugs. This review covers a number of scalable protocols for isolating and purifying exosomes from human, cow, and horse milk. Additionally, it considers passive and active methods for drug loading into exosomes, as well as methods for modifying and functionalizing the surface of milk exosomes with specific molecules for more efficient and specific delivery to target cells. In addition, the review considers various approaches to visualize exosomes and determine cellular localization and bio-distribution of loaded drug molecules in tissues. In conclusion, we outline new challenges for studying milk exosomes, a new generation of targeted delivery agents.
Collapse
Affiliation(s)
- Anna M Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Anastasia P Paramonik
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Sergey S Sedykh
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy A Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
7
|
Liu X, Li Y, Chen C, Dong J, Zhou J, Tong D, Wang L, Gao X, Kang X. Exosomal EphA2 promotes tumor metastasis of triple-negative breast cancer by damaging endothelial barrier. Clin Exp Metastasis 2023; 40:105-116. [PMID: 36380015 DOI: 10.1007/s10585-022-10194-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Many evidences show that exosomes play an important role in cancer development, invasion and metastasis. This study is based on the need to explore exosomal protein that promote breast cancer metastasis. We found that tyrosine kinase EphA2 was enriched in Triple-negative breast cancer -derived exosomes and it could disrupt the endothelial monolayer barrier through downregulating tight junction proteins of endothelial cells. These mechanisms were confirmed by in vivo experiments. After periodical injection of exosomal EphA2 into mice caudal vein, we found increased vascular permeability and breast cancer metastases in distant organs, and this phenomenon decreased dramatically after exosomal EphA2 knockdown. This study provides a new mechanism of exosome promoting breast cancer metastasis and suggests a new therapeutic target for the prevention and treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Xin Liu
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yue Li
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Chunjing Chen
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jiyang Dong
- Department of Electronic Science, National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jie Zhou
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Dandan Tong
- School of Medicine, Huaqiao University, Xiamen, 362021, Fujian, China
| | - Lei Wang
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xiang Gao
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Xinmei Kang
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
8
|
Shefer A, Yalovaya A, Tamkovich S. Exosomes in Breast Cancer: Involvement in Tumor Dissemination and Prospects for Liquid Biopsy. Int J Mol Sci 2022; 23:8845. [PMID: 36012109 PMCID: PMC9408748 DOI: 10.3390/ijms23168845] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 12/03/2022] Open
Abstract
In women, breast cancer (BC) is the most commonly diagnosed cancer (24.5%) and the leading cause of cancer death (15.5%). Understanding how this heterogeneous disease develops and the confirm mechanisms behind tumor progression is of utmost importance. Exosomes are long-range message vesicles that mediate communication between cells in physiological conditions but also in pathology, such as breast cancer. In recent years, there has been an exponential rise in the scientific studies reporting the change in morphology and cargo of tumor-derived exosomes. Due to the transfer of biologically active molecules, such as RNA (microRNA, long non-coding RNA, mRNA, etc.) and proteins (transcription factors, enzymes, etc.) into recipient cells, these lipid bilayer 30-150 nm vesicles activate numerous signaling pathways that promote tumor development. In this review, we attempt to shed light on exosomes' involvement in breast cancer pathogenesis (including epithelial-to-mesenchymal transition (EMT), tumor cell proliferation and motility, metastatic processes, angiogenesis stimulation, and immune system repression). Moreover, the potential use of exosomes as promising diagnostic biomarkers for liquid biopsy of breast cancer is also discussed.
Collapse
Affiliation(s)
- Aleksei Shefer
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Alena Yalovaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Svetlana Tamkovich
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
9
|
Chernyshev VS, Chuprov‐Netochin RN, Tsydenzhapova E, Svirshchevskaya EV, Poltavtseva RA, Merdalimova A, Yashchenok A, Keshelava A, Sorokin K, Keshelava V, Sukhikh GT, Gorin D, Leonov S, Skliar M. Asymmetric depth-filtration: A versatile and scalable method for high-yield isolation of extracellular vesicles with low contamination. J Extracell Vesicles 2022; 11:e12256. [PMID: 35942823 PMCID: PMC9451526 DOI: 10.1002/jev2.12256] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/24/2022] Open
Abstract
We developed a novel asymmetric depth filtration (DF) approach to isolate extracellular vesicles (EVs) from biological fluids that outperforms ultracentrifugation and size-exclusion chromatography in purity and yield of isolated EVs. By these metrics, a single-step DF matches or exceeds the performance of multistep protocols with dedicated purification procedures in the isolation of plasma EVs. We demonstrate the selective transit and capture of biological nanoparticles in asymmetric pores by size and elasticity, low surface binding to the filtration medium, and the ability to cleanse EVs held by the filter before their recovery with the reversed flow all contribute to the achieved purity and yield of preparations. We further demonstrate the method's versatility by applying it to isolate EVs from different biofluids (plasma, urine, and cell culture growth medium). The DF workflow is simple, fast, and inexpensive. Only standard laboratory equipment is required for its implementation, making DF suitable for low-resource and point-of-use locations. The method may be used for EV isolation from small biological samples in diagnostic and treatment guidance applications. It can also be scaled up to harvest therapeutic EVs from large volumes of cell culture medium.
Collapse
Affiliation(s)
- Vasiliy S. Chernyshev
- Skolkovo Institute of Science and TechnologyMoscowRussian Federation
- School of Biological and Medical PhysicsMoscow Institute of Physics and TechnologyDolgoprudnyMoscow RegionRussian Federation
| | - Roman N. Chuprov‐Netochin
- School of Biological and Medical PhysicsMoscow Institute of Physics and TechnologyDolgoprudnyMoscow RegionRussian Federation
| | - Ekaterina Tsydenzhapova
- School of Biological and Medical PhysicsMoscow Institute of Physics and TechnologyDolgoprudnyMoscow RegionRussian Federation
| | | | - Rimma A. Poltavtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. KulakovMinistry of Healthcare of the Russian FederationMoscowRussian Federation
| | | | - Alexey Yashchenok
- Skolkovo Institute of Science and TechnologyMoscowRussian Federation
| | | | | | - Varlam Keshelava
- Institute for Biological Instrumentation RASPushchinoRussian Federation
| | - Gennadiy T. Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. KulakovMinistry of Healthcare of the Russian FederationMoscowRussian Federation
| | - Dmitry Gorin
- Skolkovo Institute of Science and TechnologyMoscowRussian Federation
| | - Sergey Leonov
- School of Biological and Medical PhysicsMoscow Institute of Physics and TechnologyDolgoprudnyMoscow RegionRussian Federation
| | - Mikhail Skliar
- Department of Chemical EngineeringUniversity of UtahSalt Lake CityUTUSA
- The Nano Institute of UtahUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
10
|
He J, Xi N, Han Z, Luo W, Shen J, Wang S, Li J, Guo Z, Cheng H. The Role of Liquid Biopsy Analytes in Diagnosis, Treatment and Prognosis of Colorectal Cancer. Front Endocrinol (Lausanne) 2022; 13:875442. [PMID: 35846270 PMCID: PMC9279561 DOI: 10.3389/fendo.2022.875442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract worldwide and is a serious threat to human life and health. CRC occurs and develops in a multi-step, multi-stage, and multi-gene process, in which abnormal gene expression plays an important role. CRC is currently diagnosed via endoscopy combined with tissue biopsy. Compared with tissue biopsy, liquid biopsy technology has received increasingly more attention and applications in the field of molecular detection due to its non-invasive, safe, comprehensive, and real-time dynamic nature. This review article discusses the application and limitations of current liquid biopsy analytes in the diagnosis, treatment, and prognosis of CRC, as well as directions for their future development.
Collapse
Affiliation(s)
- JinHua He
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - NaiTe Xi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - ZePing Han
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - WenFeng Luo
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jian Shen
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - ShengBo Wang
- Department of Gastroenterology, Central Hospital of Panyu District, Guangzhou, China
| | - JianHao Li
- Institute of Cardiovascular Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - ZhongHui Guo
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - HanWei Cheng
- Central Laboratory of Panyu Central Hospital, Guangzhou Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
11
|
Kirkemo LL, Elledge SK, Yang J, Byrnes JR, Glasgow JE, Blelloch R, Wells JA. Cell-surface tethered promiscuous biotinylators enable comparative small-scale surface proteomic analysis of human extracellular vesicles and cells. eLife 2022; 11:73982. [PMID: 35257663 PMCID: PMC8983049 DOI: 10.7554/elife.73982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
Characterization of cell surface proteome differences between cancer and healthy cells is a valuable approach for the identification of novel diagnostic and therapeutic targets. However, selective sampling of surface proteins for proteomics requires large samples (>10e6 cells) and long labeling times. These limitations preclude analysis of material-limited biological samples or the capture of rapid surface proteomic changes. Here, we present two labeling approaches to tether exogenous peroxidases (APEX2 and HRP) directly to cells, enabling rapid, small-scale cell surface biotinylation without the need to engineer cells. We used a novel lipidated DNA-tethered APEX2 (DNA-APEX2), which upon addition to cells promoted cell agnostic membrane-proximal labeling. Alternatively, we employed horseradish peroxidase (HRP) fused to the glycan-binding domain of wheat germ agglutinin (WGA-HRP). This approach yielded a rapid and commercially inexpensive means to directly label cells containing common N-Acetylglucosamine (GlcNAc) and sialic acid glycans on their surface. The facile WGA-HRP method permitted high surface coverage of cellular samples and enabled the first comparative surface proteome characterization of cells and cell-derived small extracellular vesicles (EVs), leading to the robust quantification of 953 cell and EV surface annotated proteins. We identified a newly recognized subset of EV-enriched markers, as well as proteins that are uniquely upregulated on Myc oncogene-transformed prostate cancer EVs. These two cell-tethered enzyme surface biotinylation approaches are highly advantageous for rapidly and directly labeling surface proteins across a range of material-limited sample types.
Collapse
Affiliation(s)
- Lisa L Kirkemo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Susanna K Elledge
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Jiuling Yang
- Department of Urology, University of California, San Francisco, San Francisco, United States
| | - James R Byrnes
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Jeff E Glasgow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| | - Robert Blelloch
- Department of Urology, University of California, San Francisco, San Francisco, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
12
|
Plasma Exosomes of Patients with Breast and Ovarian Tumors Contain an Inactive 20S Proteasome. Molecules 2021; 26:molecules26226965. [PMID: 34834058 PMCID: PMC8619627 DOI: 10.3390/molecules26226965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 01/03/2023] Open
Abstract
Exosomes are directly involved in governing of physiological and pathological conditions of an organism through the transfer of information from producing to receiving cells. It can be assumed that exosomes are one of the key players of tumor dissemination since they are very stable and small enough to penetrate from various tissues into biological fluids and then back, thus interacting with tissue target cells. We evaluated the enzymatic activity and the level of 20S proteasome in tissue and exosomes of healthy females (n = 39) and patients with ovarian (n = 50) and breast (n = 108) tumors to reveal the critical role of exosomal cargo in the mediation of different types of metastases. Exosomes from plasma and ascites were isolated and characterized in according to International Society for Extracellular Vesicles guidelines. The level of 20S proteasome in tissue and exosomes was determined using Western blot analysis. Chymotrypsin- and caspase-like (ChTL and CL, respectively) peptidase activities of the proteasomes were determined using fluorogenic Suc-LLVY-AMC and Cbz-LLG-AMC substrates, respectively. We observed increased levels of 20S proteasome in ovarian cancer tissue and luminal B subtype breast cancer tissue as well as in plasma exosomes from cancer patients. Moreover, the level of the 20S proteasome in plasma exosomes and ascites exosomes in patients with ovarian tumors is comparable and higher in ovarian cancer patients with low volume ascites than in patients with moderate and high-volume ascites. We also found increased ChTL and CL activities in breast cancer and ovarian cancer tissues, as well as in peritoneal metastases in ovarian cancer, while proteasomal activity in exosomes from plasma of healthy females and all patients, as well as from ascites of ovarian tumor patients were lower than detection limit of assay. Thus, regardless of the type of tumor metastasis (lymphogenous or peritoneal), the exosomes of cancer patients were characterized by an increased level of 20S proteasome, which do not exhibit enzymatic activity.
Collapse
|
13
|
Dynamic Landscape of Extracellular Vesicle-Associated Proteins Is Related to Treatment Response of Patients with Metastatic Breast Cancer. MEMBRANES 2021; 11:membranes11110880. [PMID: 34832109 PMCID: PMC8619728 DOI: 10.3390/membranes11110880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the leading cause of cancer death in women. The majority of these deaths are due to disease metastasis, in which cancer cells disseminate to multiple organs and disrupt vital physiological functions. It is widely accepted that breast cancer cells secrete extracellular vesicles (EVs), which contain dynamic molecular cargo that act as versatile mediators of intercellular communication. Therefore, Evs. secreted by breast cancer cells could be involved in the development of metastatic disease and resistance to treatment. Moreover, changes in EV cargo could reflect the effects of therapy on their parent tumor cells. The aim of this feasibility study was to quantitatively profile the proteomes of Evs. isolated from blood samples taken from treatment sensitive and resistant metastatic breast cancer patients to identify proteins associated with responses. Three serial blood samples were collected from three patients with metastatic breast cancer receiving systemic therapy including a responder, a non-responder, and a mixed-responder. Evs. were isolated from plasma using size exclusion chromatography and their protein cargo was prepared for tandem mass tag (TMT)-labelling and quantitative analyses using two-dimensional high-performance liquid chromatography followed by tandem mass spectrometry. After filtering, we quantitatively identified 286 proteins with high confidence using a q value of 0.05. Of these, 149 were classified as EV associated candidate proteins and 137 as classical, high abundant plasma proteins. After comparing EV protein abundance between the responder and non-responder, we identified 35 proteins with unique de-regulated abundance patterns that was conserved at multiple time points. We propose that this proof-of-concept approach can be used to identify proteins which have potential as predictors of metastatic breast cancer response to treatment.
Collapse
|
14
|
Li D, Lai W, Fan D, Fang Q. Protein biomarkers in breast cancer-derived extracellular vesicles for use in liquid biopsies. Am J Physiol Cell Physiol 2021; 321:C779-C797. [PMID: 34495763 DOI: 10.1152/ajpcell.00048.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most common malignant disease in women worldwide. Early diagnosis and treatment can greatly improve the management of breast cancer. Liquid biopsies are becoming convenient detection methods for diagnosing and monitoring breast cancer due to their noninvasiveness and ability to provide real-time feedback. A range of liquid biopsy markers, including circulating tumor proteins, circulating tumor cells, and circulating tumor nucleic acids, have been implemented for breast cancer diagnosis and prognosis, with each having its own advantages and limitations. Circulating extracellular vesicles are messengers of intercellular communication that are packed with information from mother cells and are found in a wide variety of bodily fluids; thus, they are emerging as ideal candidates for liquid biopsy biomarkers. In this review, we summarize extracellular vesicle protein markers that can be potentially used for the early diagnosis and prognosis of breast cancer or determining its specific subtypes.
Collapse
Affiliation(s)
- Dan Li
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Wenjia Lai
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Di Fan
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Qiaojun Fang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China.,Sino-Danish Center for Education and Research, Beijing, People's Republic of China
| |
Collapse
|
15
|
AlHumaidi RB, Fayed B, Sharif SI, Noreddin A, Soliman SSM. Role of Exosomes in Breast Cancer Management: Evidence-Based Review. Curr Cancer Drug Targets 2021; 21:666-675. [PMID: 34077346 DOI: 10.2174/1568009621666210601115707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/09/2022]
Abstract
The field of cancer research has massively grown in recent decades, leading to a better understanding of the underlying causes and greatly improved the therapeutic approaches. Breast cancer (BC) is the third leading cause of mortality among all cancers and the most common malignant disease in women worldwide, representing one in four cancers in women. The crosstalk between cancer cells and the surrounding microenvironment is crucial for tumor progression and metastatic process. Tumor cells communicate not only through classical paracrine signaling mechanisms, including cytokines, chemokines, growth factors, but also through"exosomes". Exosomes are nano-vesicles that are released by various types of cells. Over the last decade, researchers have been attracted to the role of exosomes in breast cancer. It has been proven that exosomes influence major tumor-related pathways, including invasion, migration, epithelial-to-mesenchymal transition (EMT), metastasis, and drug resistance. Additionally, exosomes play important roles in clinical applications. Several studies have demonstrated the potential applications of exosomes in cancer therapy and diagnosis. Furthermore, exosomes have been engineered to function as nano-delivery systems of chemotherapeutic drugs. They can also be designed as vaccines to trigger the patient's immune system. This review discusses the recent progress regarding the use of exosomes as drug delivery systems, therapeutic agents, biomarkers, and vaccines against breast cancer.
Collapse
Affiliation(s)
- Razan B AlHumaidi
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Suleiman I Sharif
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Ayman Noreddin
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| |
Collapse
|
16
|
Choi ES, Faruque HA, Kim JH, Kim KJ, Choi JE, Kim BA, Kim B, Kim YJ, Woo MH, Park JY, Hur K, Lee MY, Kim DS, Lee SY, Kim E. CD5L as an Extracellular Vesicle-Derived Biomarker for Liquid Biopsy of Lung Cancer. Diagnostics (Basel) 2021; 11:diagnostics11040620. [PMID: 33808296 PMCID: PMC8067192 DOI: 10.3390/diagnostics11040620] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 01/18/2023] Open
Abstract
Cancer screening and diagnosis can be achieved by analyzing specific molecules within serum-derived extracellular vesicles (EVs). This study sought to profile EV-derived proteins to identify potential lung cancer biomarkers. EVs were isolated from 80 serum samples from healthy individuals and cancer patients via polyethylene glycol (PEG)-based precipitation and immunoaffinity separation using antibodies against CD9, CD63, CD81, and EpCAM. Proteomic analysis was performed using 2-D gel electrophoresis and matrix-assisted laser desorption ionization–time-of-flight mass spectrometry (MALDI–TOF MS). The expression of proteins that were differentially upregulated in the EVs or tissue of lung cancer samples was validated by Western blotting. The area under the curve (AUC) was calculated to assess the predictability of each differentially expressed protein (DEP) for lung cancer. A total of 55 upregulated protein spots were selected, seven of which (CD5L, CLEC3B, ITIH4, SERFINF1, SAA4, SERFINC1, and C20ORF3) were found to be expressed at high levels in patient-derived EVs by Western blotting. Meanwhile, only the expression of EV CD5L correlated with that in cancer tissues. CD5L also demonstrated the highest AUC value (0.943) and was found to be the core regulator in a pathway related to cell dysfunction. Cumulatively, these results show that EV-derived CD5L may represent a potential biomarker—detected via a liquid biopsy—for the noninvasive diagnosis of lung cancer.
Collapse
Affiliation(s)
- Eun-Sook Choi
- Division of Bi-Fusion Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Dague 42988, Korea; (E.-S.C.); (H.A.F.)
| | - Hasan Al Faruque
- Division of Bi-Fusion Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-jungangdaero 333, Dague 42988, Korea; (E.-S.C.); (H.A.F.)
| | - Jung-Hee Kim
- Division of Electronic Information System Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-Jungangdaero 333, Dague 42988, Korea;
| | - Kook Jin Kim
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Jin Eun Choi
- Department of Biochemistry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.E.C.); (K.H.)
| | - Bo A. Kim
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Bora Kim
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Ye Jin Kim
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Min Hee Woo
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Keun Hur
- Department of Biochemistry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.E.C.); (K.H.)
| | - Mi-Young Lee
- Department of Medical Science, Soonchunhyang University, Asan 31538, Korea;
| | - Dong Su Kim
- Genomine Research Division, Genomine Inc., Pohang Technopark, Pohang 37668, Korea; (K.J.K.); (B.A.K.); (B.K.); (Y.J.K.); (M.H.W.); (D.S.K.)
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
- Correspondence: (S.Y.L.); (E.K.); Tel.: +82-53-200-2632 (S.Y.L.); +82-53-785-2530 (E.K.)
| | - Eunjoo Kim
- Division of Electronic Information System Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Techno-Jungangdaero 333, Dague 42988, Korea;
- Correspondence: (S.Y.L.); (E.K.); Tel.: +82-53-200-2632 (S.Y.L.); +82-53-785-2530 (E.K.)
| |
Collapse
|
17
|
Yunusova NV, Zambalova EA, Patysheva MR, Kolegova ES, Afanas'ev SG, Cheremisina OV, Grigor'eva AE, Tamkovich SN, Kondakova IV. Exosomal Protease Cargo as Prognostic Biomarker in Colorectal Cancer. Asian Pac J Cancer Prev 2021; 22:861-869. [PMID: 33773551 PMCID: PMC8286660 DOI: 10.31557/apjcp.2021.22.3.861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: The aim of the study was to develop a model for predicting cancer risk in colorectal polyps’ patients (CPPs), as well as to reveal additional prognosis factors for Stage III colorectal cancer based on differences in subpopulations of tetraspanins, tetraspanin-associated and tetraspanin-non-associated proteases in blood plasma exosomes of CPPs and colorectal cancer patients (CRCPs). Methods: The subpopulations of CD151- and Tspan8-positive exosomes, the subpopulations of metalloproteinase at the surface of СD9-positive exosomes and the level of 20S proteasomes in plasma exosomes in 15 CPPs (tubulovillous adenomas) and 60 CRCPs were evaluated using flow cytometry and Western blotting. Logistic regression analysis was performed to predict cancer risk of CPPs. Results: The levels of 20S proteasomes in exosomes, MMP9+, MMP9+/MMP2+/EMMPRIN+ in CD9-positive blood plasma exosomes are associated with the risk of malignant transformation of colorectal tubulovillous adenomas. In patients with Stage III CRC, the levels of 20S proteasomes (less than 2 units) and MMP9+ subpopulations (more than 61%) in plasma exosomes are unfavorable prognostic factors for overall survival. The levels of 20S proteasomes and ADAM10+/ADAM17- subpopulations in CD9-positive blood plasma exosomes are the most significant values for predicting relapse-free survival. Conclusion: Protease cargo in CD9-positive blood plasma exosomes is prognostic biomarker for colorectal polyps and colorectal cancer.
Collapse
Affiliation(s)
- Natalia V Yunusova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.,Department of Biochemistry and Molecular Biology, Faculty of Medicine and Biology, Siberian State Medical University, Tomsk, Russia
| | - Elena A Zambalova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Marina R Patysheva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Elena S Kolegova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey G Afanas'ev
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Olga V Cheremisina
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Alina E Grigor'eva
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Svetlana N Tamkovich
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Clinical Biochemistry, V. Zelman Institute for the Medicine, Novosibirsk, Russia
| | - Irina V Kondakova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
18
|
Parada N, Romero-Trujillo A, Georges N, Alcayaga-Miranda F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J Adv Res 2021; 31:61-74. [PMID: 34194832 PMCID: PMC8240105 DOI: 10.1016/j.jare.2021.01.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/29/2020] [Accepted: 01/01/2021] [Indexed: 12/14/2022] Open
Abstract
Background Even though exosome-based therapy has been shown to be able to control the progression of different pathologies, the data revealed by pharmacokinetic studies warn of the low residence time of exogenous exosomes in circulation that can hinder the clinical translation of therapeutic exosomes. The macrophages related to the organs of the mononuclear phagocytic system are responsible primarily for the rapid clearance and retention of exosomes, which strongly limits the amount of exosomal particles available to reach the target tissue, accumulate in it and release with high efficiency its therapeutic cargo in acceptor target cells to exert the desired biological effect. Aim of review Endowing exosomes with surface modifications to evade the immune system is a plausible strategy to contribute to the suppression of exosomal clearance and increase the efficiency of their targeted content delivery. Here, we summarize the current evidence about the mechanisms underlying the recognition and sequestration of therapeutic exosomes by phagocytic cells. Also, we propose different strategies to generate 'invisible' exosomes for the immune system, through the incorporation of different anti-phagocytic molecules on the exosomes’ surface that allow increasing the circulating half-life of therapeutic exosomes with the purpose to increase their bioavailability to reach the target tissue, transfer their therapeutic molecular cargo and improve their efficacy profile. Key scientific concepts of review Macrophage-mediated phagocytosis are the main responsible behind the short half-life in circulation of systemically injected exosomes, hindering their therapeutic effect. Exosomes ‘Camouflage Cloak’ strategy using antiphagocytic molecules can contribute to the inhibition of exosomal clearance, hence, increasing the on-target effect. Some candidate molecules that could exert an antiphagocytic role are CD47, CD24, CD44, CD31, β2M, PD-L1, App1, and DHMEQ. Pre- and post-isolation methods for exosome engineering are compatible with the loading of therapeutic cargo and the expression of antiphagocytic surface molecules.
Collapse
Affiliation(s)
- Nicol Parada
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Alfonso Romero-Trujillo
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Nicolás Georges
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Francisca Alcayaga-Miranda
- School of Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| |
Collapse
|
19
|
Martini V, D'Avanzo F, Maggiora PM, Varughese FM, Sica A, Gennari A. Oncolytic virotherapy: new weapon for breast cancer treatment. Ecancermedicalscience 2020; 14:1149. [PMID: 33574894 PMCID: PMC7864690 DOI: 10.3332/ecancer.2020.1149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
The recent introduction of viruses as a weapon against cancer can be regarded as one of the most intriguing approaches in the context of precision medicine. The role of immune checkpoint inhibitors has been extensively studied in early and advanced cancer stages, with extraordinary results. Although there is a good tolerability profile, especially when compared with conventional chemotherapy, severe immune-related adverse events have emerged as a potential limitation. Moreover, there are still treatment-resistant cases and thus further treatment options need to be implemented. Several in vitro and in vivo studies have been conducted and are ongoing to develop oncolytic viruses (OVs) as a tool to modulate the immune system response. OVs are attenuated viruses that can kill cancer cells after having infected them, producing microenvironment remodelling and antitumour immune response. The potential of oncolytic virotherapy is to contrast the absence of T cell infiltrates, converting ‘cold’ tumours into ‘hot’ ones, thus improving the performance of the immune system. Breast cancer, the second most common cause of cancer-related deaths among women, is considered a ‘cold’ tumour. In this context, oncolytic virotherapy might well be considered as a promising strategy. This review summarises the current status, clinical applications and future development of OVs, focusing on breast cancer treatment.
Collapse
Affiliation(s)
- Veronica Martini
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0887-4082
| | - Francesca D'Avanzo
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Paola Maria Maggiora
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy
| | - Feba Maria Varughese
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A Avogadro 28100, Italy.,Department of Inflammation and Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano (MI) 20089, Italy.,https://orcid.org/0000-0002-8342-7442
| | - Alessandra Gennari
- Division of Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara 13100, Italy.,Center for Translational Research on Autoimmune & Allergic Diseases - CAAD, Novara 28100, Italy.,https://orcid.org/0000-0002-0928-2281
| |
Collapse
|
20
|
Tutanov O, Proskura K, Kamyshinsky R, Shtam T, Tsentalovich Y, Tamkovich S. Proteomic Profiling of Plasma and Total Blood Exosomes in Breast Cancer: A Potential Role in Tumor Progression, Diagnosis, and Prognosis. Front Oncol 2020. [DOI: 10.3389/fonc.2020.580891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
21
|
Matsumoto Y, Kano M, Murakami K, Toyozumi T, Suito H, Takahashi M, Sekino N, Shiraishi T, Kamata T, Ryuzaki T, Hirasawa S, Kinoshita K, Matsubara H. Tumor-derived exosomes influence the cell cycle and cell migration of human esophageal cancer cell lines. Cancer Sci 2020; 111:4348-4358. [PMID: 32969511 PMCID: PMC7734159 DOI: 10.1111/cas.14660] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/22/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023] Open
Abstract
Our laboratory previously reported the usefulness as biomarkers of exosomes in the plasma of esophageal squamous cell carcinoma (ESCC) patients. However, the influence of tumor‐derived exosomes on the tumor itself and underlying mechanisms remain unclear. We here report changes in the phenotype and gene expression when cancer cells exist in an environment with tumor‐derived exosomes. The exosomes were isolated from the culture medium of human ESCC cells (TE2, T.Tn) by ultracentrifugation; cell proliferation assay, wound‐healing assay, and fluorescence imaging of the cell cycle were performed to clarify the phenotypic changes in the high concentration of tumor‐derived exosomes. Gene expression changes were also assessed by mRNA microarray, and the data were analyzed by gene set enrichment analysis (GSEA). The data revealed that the proliferation of both TE2 and T.Tn was inhibited, and cell migration ability was upregulated in the exosome exposure group (P < .05). Fluorescence imaging using a fluorescent ubiquitination‐based cell cycle indicator expressing ESCC cells revealed that the ratio of G1‐phase cells was significantly increased in the exosome exposure group (P < .05). Findings of the GSEA clarified that high‐density exposure of cancer‐derived exosomes to their parent cancer cells downregulated the expression of genes related to cell proliferation and cell cycle, and upregulated the expression of genes related to actin filament length and extracellular structure organization. In conclusion, an environment of high‐density tumor‐derived exosomes induces changes in the gene expression and phenotype of tumor cells and may lead to tumor progression or malignant transformation.
Collapse
Affiliation(s)
- Yasunori Matsumoto
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masayuki Kano
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kentaro Murakami
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takeshi Toyozumi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hiroshi Suito
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masahiko Takahashi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nobufumi Sekino
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tadashi Shiraishi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toshiki Kamata
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takahiro Ryuzaki
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Soichiro Hirasawa
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuya Kinoshita
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
22
|
Konoshenko M, Sagaradze G, Orlova E, Shtam T, Proskura K, Kamyshinsky R, Yunusova N, Alexandrova A, Efimenko A, Tamkovich S. Total Blood Exosomes in Breast Cancer: Potential Role in Crucial Steps of Tumorigenesis. Int J Mol Sci 2020; 21:E7341. [PMID: 33027894 PMCID: PMC7582945 DOI: 10.3390/ijms21197341] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/19/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022] Open
Abstract
Exosomes are crucial players in cell-to-cell communication and are involved in tumorigenesis. There are two fractions of blood circulating exosomes: free and cell-surface-associated. Here, we compared the effect of total blood exosomes (contain plasma exosomes and blood cell-surface-associated exosomes) and plasma exosomes from breast cancer patients (BCPs, n = 43) and healthy females (HFs, n = 35) on crucial steps of tumor progression. Exosomes were isolated by ultrafiltration, followed by ultracentrifugation, and characterized by cryo-electron microscopy (cryo-EM), nanoparticle tracking analysis, and flow cytometry. Cryo-EM revealed a wider spectrum of exosome morphology with lipid bilayers and vesicular internal structures in the HF total blood in comparison with plasma. No differences in the morphology of both exosomes fractions were detected in BCP blood. The plasma exosomes and total blood exosomes of BCPs had different expression levels of tumor-associated miR-92a and miR-25-3p, induced angiogenesis and epithelial-to-mesenchymal transition (EMT), and increased the number of migrating pseudo-normal breast cells and the total migration path length of cancer cells. The multidirectional effects of HF total blood exosomes on tumor dissemination were revealed; they suppress the angiogenesis and total migration path length of MCF10A, but stimulate EMT and increase the number of migrating MCF10A and the total path length of SKBR3 cells. In addition, HF plasma exosomes enhance the metastasis-promoting properties of SKBR3 cells and stimulate angiogenesis. Both cell-free and blood cell-surface-associated exosomes are involved in the crucial stages of carcinogenesis: the initiation of EMT and the stimulation of proliferation, cell migration, and angiogenesis. Thus, for the estimation of the diagnostic/prognostic significance of circulating exosomes in the blood of cancer patients more correctly, the total blood exosomes, which consist of plasma exosomes and blood cell-surface-associated exosomes should be used.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
| | - Georgy Sagaradze
- Medical Research and Education Center, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.S.); (A.E.)
| | - Evgeniya Orlova
- N.N. Blokhin Cancer Research Center” of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (E.O.); (A.A.)
| | - Tatiana Shtam
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Gatchina, Russia;
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia;
| | - Ksenia Proskura
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
- Novosibirsk Regional Clinical Oncological Dispensary, 630108 Novosibirsk, Russia
| | - Roman Kamyshinsky
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia;
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow region, Russia
- Shubnikov Institute of Crystallography of Federal Scientific Research Centre, “Crystallography and Photonics” of Russian Academy of Sciences, 119333 Moscow, Russia
| | - Natalia Yunusova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Science, 634050 Tomsk, Russia;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Biology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Antonina Alexandrova
- N.N. Blokhin Cancer Research Center” of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (E.O.); (A.A.)
| | - Anastasia Efimenko
- Medical Research and Education Center, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.S.); (A.E.)
| | - Svetlana Tamkovich
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
- Department of Molecular Biology and Biotechnology, Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|