1
|
Colangelo T, Mazzarelli F, Cuttano R, Dama E, Melocchi V, Afanga MK, Perrone RM, Graziano P, Bianchi F. Unveiling the origin and functions of diagnostic circulating microRNAs in lung cancer. Br J Cancer 2025; 132:947-956. [PMID: 40185877 DOI: 10.1038/s41416-025-02982-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Circulating microRNAs (c-miRs) were shown to be effective biomarkers for lung cancer early detection. However, the understanding of c-miRs origin and their biological functions still remains elusive. METHODS We analysed miRNA expression in a large panel of lung cancer (LC) and hematopoietic cell lines (N = 252; CCLE database) coupled with c-miR profile of a large cohort of serum samples (N = 975), from high-risk subjects underwent annual LD-CT for 5 years. Furthermore, we examined intracellular and extracellular miR-29a-3p/223-3p expression profile in lung adenocarcinoma (LUAD) tissues, in matched serum samples and in LC and stromal cell lines. Lastly, through the modulation of expression of selected c-miRs by using mimic (OE) or antisense microRNA (KD), we explored their impact on lung cancer transcriptome and cancer and immune phenotypes. RESULTS Here, we investigated the origin of an extensively validated 13 c-miRs signature diagnostics for asymptomatic lung cancer (LC) in high-risk subjects (smokers, >20 packs/y; >50 y old). Overall, we found a mixed origin of these c-miRs, originating both from tumour cells and the tumour microenvironment (TME). Intriguingly, we revealed that circulating miR-29a-3p and miR-223-3p are abundantly released from LC epithelial cells and immune cells, respectively. In particular, we found that miR-223-3p triggered several lung cancer related phenotypes such as invasion, migration and tumour-promoting inflammation. CONCLUSIONS Our study highlights a mixed tumour epithelial and stroma-associated origin of LC c-miRs with new evidences on the multifaceted role of miR-223-3p in LC pathogenesis and immune modulation.
Collapse
Affiliation(s)
- Tommaso Colangelo
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesco Mazzarelli
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Roberto Cuttano
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Elisa Dama
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Valentina Melocchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Miriam Kuku Afanga
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Rosa Maria Perrone
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Paolo Graziano
- Unit of Pathology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Roma, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
2
|
Shirzad S, Eterafi M, Karimi Z, Barazesh M. MicroRNAs involved in colorectal cancer, a rapid mini-systematic review. BMC Cancer 2025; 25:934. [PMID: 40413456 DOI: 10.1186/s12885-025-14343-1] [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: 11/29/2024] [Accepted: 05/16/2025] [Indexed: 05/27/2025] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) involves the uncontrolled proliferation of glandular epithelial cells in the colon or rectum. The high mortality rate associated with CRC has driven extensive research into innovative diagnostic and therapeutic strategies. Among these, microRNAs (miRNA) have gained attention for their crucial role in regulating various cellular processes that contribute to the initiation, progression, and metastasis of CRC. METHOD This systematic review aimed to assess the roles of various miRNAs in CRC by analyzing multiple studies. The PICO framework was followed to structure the study regarding miRNA involved in CRC development and progression compared to normal cases. The outcomes were measured according miRNAs impact on CRC progression, survival rates, and treatment response. Systematic review of studies published from 2000 to November 2023 were included. Data were collected from prominent databases, including Google Scholar, PubMed, ScienceDirect, Irandoc, SID, and Magiran, covering studies from 2000 to November 2023. Studies were managed using EndNote for citation management, and duplicates were removed. The remaining studies were evaluated based on predefined inclusion and exclusion criteria. RESULTS In our review, we categorized 28 miRNAs based on their potential tumor suppressor or oncogenic effects in CRC progression. Among them, 14 miRNAs were highlighted as important based on the assessment using TCGA data, with miR-200a also showing a significant effect on patient survival. CONCLUSION This study compiled and analyzed validated miRNAs associated with CRC progression. The findings suggest the potential of these miRNAs as non-invasive biomarkers, which may be used alone or in combination with traditional tumor markers for improved diagnostic and prognostic applications in CRC. This review contributes novel insights by updating the current understanding and offering a comprehensive evaluation of miRNAs in CRC.
Collapse
Affiliation(s)
- Sogol Shirzad
- Students Research Committee, Gerash University of Medical Sciences, Gerash, Iran
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran
| | - Majid Eterafi
- Students Research Committee, Gerash University of Medical Sciences, Gerash, Iran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zeinab Karimi
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran.
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran.
| | - Mahdi Barazesh
- Medical Biotechnology Group, Gerash University of Medical Sciences, Gerash, Iran
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
3
|
Islam MKB, Marcus RK. Effects of packing density and adsorption conditions on extracellular vesicle dynamic binding capacities for capillary-channeled polymer (C-CP) fiber columns. J Chromatogr A 2025; 1755:466068. [PMID: 40403650 DOI: 10.1016/j.chroma.2025.466068] [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: 03/12/2025] [Revised: 05/16/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
Extracellular vesicles (EVs) are membrane-bound nanoparticles (50-1000 nm) secreted by all cell types and play critical roles in various biological processes. Among these, exosomes, a smaller subset of EVs, have attracted considerable interest due to their potential applications in diagnostics and therapeutics. However, conventional EV isolation methods are often limited by inefficiencies in processing time, recovery, and scalability. Hydrophobic interaction chromatography utilizing capillary-channeled polymer (CCP) fiber stationary phases offers a promising alternative, enabling rapid (<15 min), cost-effective (∼$5 per column) EV isolation with high loading capacities (∼1010-10¹² particles) and minimal sample pre-processing. Despite these advantages, achieving high-throughput EV isolation for larger-scale applications using the CCP fiber platform is the present challenge. To this end, further optimization of stationary phase packing and adsorption conditions is necessary to maximize the available binding surface area in the current microbore column format. This study systematically investigates the influence of interstitial fraction (i.e. packing density) in polyester (PET) CCP fiber columns on the dynamic binding capacity (DBC) of EVs isolated from human urine using a high-performance liquid chromatography platform. Microbore columns (0.76 mm i.d. × 300 mm) packed with PET CCP fibers in both an eight-channel (PET-8) and a novel trilobal (PET-Y) configuration were evaluated using breakthrough curves and frontal analysis. The results reveal that lower packing densities correlate with higher mass- and surface area-based EV binding capacities, with a maximum DBCs of 2.86 × 10¹³ EVs g-1 fiber and 1.22 × 10¹⁴ EVs m⁻² fiber achieved in <2 min of sample loading. Under optimum conditions, surface utilization of >50 % is realized. These results establish a framework for optimizing CCP fiber-based platforms to enhance EV capture efficiency, facilitating the development of scalable EV isolation techniques for biomedical research and therapeutic applications.
Collapse
Affiliation(s)
- Md Khalid Bin Islam
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC 29634-0973, USA
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC 29634-0973, USA.
| |
Collapse
|
4
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
5
|
Abida, Alhuthali HM, Alshehri JM, Alkathiri A, Almaghrabi ROM, Alsaeed SS, Albebi SAH, Almethn RM, Alfuraydi BA, Alharbi SB, Kamal M, Imran M. Exosomes in infectious diseases: insights into leishmaniasis pathogenesis, immune modulation, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4913-4931. [PMID: 39702600 DOI: 10.1007/s00210-024-03702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Leishmaniasis continues to be a critical international health issue due to the scarcity of efficient treatment and the development of drug tolerance. New developments in the research of extracellular vesicles (EVs), especially exosomes, have revealed novel disease management approaches. Exosomes are small vesicles that transport lipids, nucleic acids, and proteins in cell signalling. Its biogenesis depends on several cellular processes, and their functions in immune response, encompassing innate and adaptive immunity, underline their function in the pathogen-host interface. Exosomes play a significant role in the pathogenesis of some parasitic infections, especially Leishmaniasis, by helping parasites escape host immunity and promote disease progression. This article explains that in the framework of parasitic diseases, exosomes can act as master regulators that define the pathogenesis of the disease, as illustrated by the engagement of exosomes in the Leishmaniasis parasite and immune escape processes. Based on many published articles on Leishmaniasis, this review aims to summarize the biogenesis of exosomes, the properties of the cargo in exosomes, and the modulation of immune responses. We delve deeper into the prospect of using exosomes for the therapy of Leishmaniasis based on the possibility of using these extracellular vesicles for drug delivery and as diagnostic and prognostic biomarkers. Lastly, we focus on the recent research perspectives and future developments, underlining the necessity to continue the investigation of exosome-mediated approaches in Leishmaniasis treatment. Thus, this review intends to draw attention to exosomes as a bright new perspective in the battle against this disabling affliction.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Jawaher Mohammad Alshehri
- Optometry Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Afnan Alkathiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Ruba Omar M Almaghrabi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | | | | | | | | | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
6
|
Liu C, Luo Y, Zhou H, Lin M, Zang D, Chen J. Immune cell-derived exosomal non-coding RNAs in tumor microenvironment: Biological functions and potential clinical applications. Chin J Cancer Res 2025; 37:250-267. [PMID: 40353080 PMCID: PMC12062983 DOI: 10.21147/j.issn.1000-9604.2025.02.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
The intricate interactions between immune cells and tumors exert a profound influence on cancer progression and therapeutic efficacy. Within the tumor microenvironment, exosomes have emerged as pivotal mediators of intercellular communication, with their cargo of non-coding RNAs (ncRNAs) serving as key regulatory elements. This review examines the multifaceted roles of immune cell-derived exosomal ncRNAs in tumor biology. The involvement of various immune cells, including T cells, B cells, natural killer cells, macrophages, neutrophils, and myeloid-derived suppressor cells, in utilizing exosomal ncRNAs to regulate tumor initiation and progression is explored. Additionally, the biogenesis and delivery mechanisms of these immune cell-derived exosomal ncRNAs are discussed, alongside their potential clinical applications in cancer.
Collapse
Affiliation(s)
- Chenguang Liu
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Yawen Luo
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Huan Zhou
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Meixi Lin
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Dan Zang
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Jun Chen
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
7
|
Wang J, Tang J, Liang A, Cui Z, Huo J, Li Q, Ke B, Yang D, Yao C. A Smart DNA Network-Based Diagnostic System for Enrichment and Detection of Circulating Tumor Cells in Cancer Liquid Biopsy. Anal Chem 2025; 97:8065-8072. [PMID: 40185686 DOI: 10.1021/acs.analchem.5c00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as critical biomarkers in liquid biopsy for noninvasive tumor diagnosis and real-time monitoring of cancer progression. However, the isolation of CTCs is often required before detection due to their ultralow abundance in peripheral blood. These isolation processes are typically time-consuming and prone to cell loss, which limits the utility of CTC-based liquid biopsy. Herein, we present a DNA network-based diagnostic system that enables specific recognition, selective enrichment, and accurate detection of CTCs directly from blood samples. The DNA network comprises ultralong DNA chains embedded with polyvalent aptamers and fluorescence detection modules. The polyvalent aptamers selectively bind to the epithelial cell adhesion molecule (EpCAM) on a CTC membrane, facilitating their enrichment through base pairing-driven DNA network formation. This system semiquantitatively detects the expression level of cancer-associated microRNA within CTCs using ratiometric fluorescence imaging based on the chemical assembly of two fluorescence modules. In clinical blood samples, this diagnostic system achieves 100% precision and 96% accuracy in distinguishing breast cancer patients from healthy donors, highlighting its promising potential for clinical breast cancer diagnosis.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Jianpu Tang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Aiqi Liang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Zhen Cui
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Jiale Huo
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Qian Li
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Bin Ke
- Department of Gastric Surgery, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Dayong Yang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Chi Yao
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| |
Collapse
|
8
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
9
|
Neophytou K, Martínez-Ugalde I, Fenton T, Robertson E, Strachan LJ, Harcus Y, Naar CM, Wright D, Price DRG, White R, Evans MJ, Bermúdez-Barrientos JR, Li H, Maizels RM, Aroian RV, Nisbet AJ, Abreu-Goodger C, Buck AH. A non-vesicular Argonaute protein is transmitted from nematode to mouse and is important for parasite survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646544. [PMID: 40236219 PMCID: PMC11996448 DOI: 10.1101/2025.04.01.646544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Argonautes are ancient proteins with well-characterised functions in cell-autonomous gene regulation and genome defense but less clear roles in non-cell-autonomous processes. Extracellular Argonautes have been reported across plants, animals and protozoa yet their biochemical and functional properties remain elusive. Here we demonstrate that an extracellular Argonaute (exWAGO) released by the rodent-infective parasitic nematode Heligmosomoides bakeri is detectable inside mouse cells during the natural infection. We further show that exWAGO is released from H.bakeri in both vesicular and non-vesicular forms that have different resistances to proteolysis, different accessibilities to antibodies and associate with different subsets of secondary siRNAs. Using recombinant exWAGO protein we demonstrate that non-vesicular exWAGO is directly internalised by mouse cells in vitro and that immunisation of mice with exWAGO confers partial protection against subsequent H. bakeri infection and generates antibodies that block exWAGO uptake into cells. Finally, we show that properties of exWAGO are conserved across Clade V nematodes that infect humans and livestock. Together this work expands the context in which Argonautes function and illuminates an RNA-binding protein as a vaccine target for parasitic nematodes.
Collapse
|
10
|
Bi W, Cao X, Li J, Gao Y, Song Y, He B. Ultrasensitive Detection of Extracellular Vesicles Based on Metal-Organic Framework DNA Biobarcodes Triggered G-Quadruplex Coupled with Rolling Circle Amplification Assay. ACS Sens 2025; 10:2136-2146. [PMID: 40048560 DOI: 10.1021/acssensors.4c03384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Extracellular vesicles (EVs), as liquid biopsy markers for accurate tumor diagnosis, are considered to hold great promise. However, effectively isolating and sensitively detecting EVs with convenience still face challenges. Herein, we propose a highly sensitive and specific platform for EV detection by integrating a metal-organic framework (MOF)-based DNA biobarcodes strategy with a rolling circle amplification (RCA)/G-quadruplex system. In this study, first, Zr-MOFs act as signal converters by comodification with DNA barcodes and antibodies, converting and amplifying the abundance of EVs into DNA barcodes. Second, the released DNA can trigger RCA, followed by G-quadruplex formation to further amplify the signal. Consequently, this approach significantly enhances the sensitivity for EV biomarker detection, achieving a low limit of detection of 100 EVs mL-1. Furthermore, the strategy offers high sensitivity, specificity, accuracy, and simplicity, highlighting its potential for clinical applications in noninvasive EV detection.
Collapse
Affiliation(s)
- Wen Bi
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaoqing Cao
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingjing Li
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
11
|
Peng J, Liu W, Tian J, Shu Y, Zhao R, Wang Y. Non-coding RNAs as key regulators of epithelial-mesenchymal transition in breast cancer. Front Cell Dev Biol 2025; 13:1544310. [PMID: 40201201 PMCID: PMC11975958 DOI: 10.3389/fcell.2025.1544310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
This study examines the critical role of non-coding RNAs (ncRNAs) in regulating epithelial-mesenchymal transition (EMT) in breast cancer, a prevalent malignancy with significant metastatic potential. EMT, wherein cancer cells acquire mesenchymal traits, is fundamental to metastasis. ncRNAs-such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)-modulate EMT by influencing gene expression and signaling pathways, affecting cancer cell migration and invasion. This review consolidates recent findings on ncRNA-mediated EMT regulation and explores their diagnostic and therapeutic potential. Specifically, miRNAs inhibit EMT-related transcription factors, while lncRNAs and circRNAs regulate gene expression through interactions with miRNAs, impacting EMT progression. Given the influence of ncRNAs on metastasis and therapeutic resistance, advancing ncRNA-based biomarkers and treatments holds promise for improving breast cancer outcomes.
Collapse
Affiliation(s)
- Jing Peng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenhui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiaju Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuncong Shu
- School of life science, Lanzhou University, Lanzhou, China
| | - Rui Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Chi H, Shi L, Gan S, Fan G, Dong Y. Innovative Applications of Nanopore Technology in Tumor Screening: An Exosome-Centric Approach. BIOSENSORS 2025; 15:199. [PMID: 40277513 PMCID: PMC12024935 DOI: 10.3390/bios15040199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
Cancer remains one of the leading causes of death worldwide. Its complex pathogenesis and metastasis pose significant challenges for early diagnosis, underscoring the urgent need for innovative and non-invasive tumor screening methods. Exosomes, small extracellular vesicles that reflect the physiological and pathological states of their parent cells, are uniquely suited for cancer liquid biopsy due to their molecular cargo, including RNA, DNA, and proteins. However, traditional methods for exosome isolation and detection are often limited by inadequate sensitivity, specificity, and efficiency. Nanopore technology, characterized by high sensitivity and single-molecule resolution, offers powerful tools for exosome analysis. This review highlights its diverse applications in tumor screening, such as magnetic nanopores for high-throughput sorting, electrochemical sensing for real-time detection, nanomaterial-based assemblies for efficient capture, and plasmon resonance for ultrasensitive analysis. These advancements have enabled precise exosome detection and demonstrated promising potential in the early diagnosis of breast, pancreatic, and prostate cancers, while also supporting personalized treatment strategies. Additionally, this review summarizes commercialized products for exosome-based cancer diagnostics and examines the technical and translational challenges in clinical applications. Finally, it discusses the future prospects of nanopore technology in advancing liquid biopsy toward clinical implementation. The continued progress of nanopore technology not only accelerates exosome-based precision medicine but also represents a significant step forward in next-generation liquid biopsy and tumor screening.
Collapse
Affiliation(s)
- Heng Chi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | - Liuxin Shi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | | | | | - Yuliang Dong
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
- BGI Research, Hangzhou 310030, China;
| |
Collapse
|
13
|
Ghufran SM, Brown ML, Beierle EA. Role of exosomes in diagnosis, prognostication, and treatment of pediatric solid tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200930. [PMID: 39895692 PMCID: PMC11783428 DOI: 10.1016/j.omton.2024.200930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cancer is the second leading cause of death in children, and solid tumors make up 30% of childhood cancers. Molecular profiling of pediatric solid tumors allows a personalized approach to therapy, but this approach mostly relies on surgical biopsy, which is invasive and carries the risk of complications. Liquid biopsy serves as a reliable alternative and a minimally invasive tool for diagnosing, prognosticating, and residual disease monitoring in childhood cancers. This review outlines the potential of exosomes as informative liquid biopsies in pediatric solid tumors. Studies highlighting the potential applications and clinical utility of exosomes and their molecular constituents as prognosticators and therapies in common childhood solid tumors, including neuroblastoma, medulloblastoma, sarcoma, and hepatoblastoma, have been overviewed. We also discuss the limitations and technical challenges of utilizing exosomes for pediatric solid tumors.
Collapse
Affiliation(s)
- Shaikh M. Ghufran
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Morgan L. Brown
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Elizabeth A. Beierle
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| |
Collapse
|
14
|
Persano F, Parodi A, Pallaeva T, Kolesova E, Zamyatnin AA, Pokrovsky VS, De Matteis V, Leporatti S, Cascione M. Atomic Force Microscopy: A Versatile Tool in Cancer Research. Cancers (Basel) 2025; 17:858. [PMID: 40075706 PMCID: PMC11899184 DOI: 10.3390/cancers17050858] [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: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The implementation of novel analytic methodologies in cancer and biomedical research has enabled the quantification of parameters that were previously disregarded only a few decades ago. A notable example of this paradigm shift is the widespread integration of atomic force microscopy (AFM) into biomedical laboratories, significantly advancing our understanding of cancer cell biology and treatment response. AFM allows for the meticulous monitoring of different parameters at the molecular and nanoscale levels, encompassing critical aspects such as cell morphology, roughness, adhesion, stiffness, and elasticity. These parameters can be systematically investigated in correlation with specific cell treatment, providing important insights into morpho-mechanical properties during normal and treated conditions. The resolution of this system holds the potential for its systematic adoption in clinics; its application could produce useful diagnostic information regarding the aggressiveness of cancer and the efficacy of treatment. This review endeavors to analyze the current literature, underscoring the pivotal role of AFM in biomedical research, especially in cancer cases, while also contemplating its prospective application in a clinical context.
Collapse
Affiliation(s)
- Francesca Persano
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
| | - Tatiana Pallaeva
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
- Federal Scientific Research Center Crystallography and Photonics, Russian Academy of Sciences, 119333 Moscow, Russia
| | - Ekaterina Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
| | - Andrey A. Zamyatnin
- Department of Biological Chemistry, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Vadim S. Pokrovsky
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (T.P.); (E.K.); (V.S.P.)
- N.N. Blokhin Medical Research Center of Oncology, 115478 Moscow, Russia
- Patrice Lumumba People’s Friendship University, 117198 Moscow, Russia
| | - Valeria De Matteis
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- Institute for Microelectronics and Microsystems (IMM), National Research Council (CNR), Via Monteroni, 73100 Lecce, Italy
| | - Stefano Leporatti
- CNR Nanotec-Istituto di Nanotecnologia, Via Monteroni, 73100 Lecce, Italy
| | - Mariafrancesca Cascione
- Mathematics and Physics Department “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (F.P.); (V.D.M.)
- Institute for Microelectronics and Microsystems (IMM), National Research Council (CNR), Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
15
|
Wang X, Yang X, Huang C, Liu T, Zang H, Gu Y, Zhang Y, Zhu X, Zhang C, Guo F, Wu S, Ding A, Yin R, Ye Q, Gao S. Tumor-derived extracellular vesicle PD-1 promotes tumor immune evasion via disruption of peripheral T cell homeostasis. Cancer Lett 2025; 612:217486. [PMID: 39864541 DOI: 10.1016/j.canlet.2025.217486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025]
Abstract
The programmed cell death 1 (PD-1)/PD-1 ligand 1 (PD-L1) axis mediates immune evasion of tumor, and targeting this axis has achieved some clinical benefits. The regulation of PD-1 expression in immune cells has been well studied. However, whether any other potential source of immune cell-expressed PD-1 exists remains unknown. Here, we report that tumor cells express PD-1 and release PD-1 in the form of extracellular vesicles, which enters T cells and suppresses T cell function via PD-L1 in vitro. In vivo, tumor cell-derived extracellular vesicle PD-1 promotes tumor growth via disrupting peripheral T cell homeostasis, showing by decreased number of T cells and impaired function of CD8+ T cells in spleens, draining lymph nodes and tumor infiltrating lymphocytes, which is restored by PD-1-targeted antibodies. Our study provides a unique and novel perspective for immune evasion of tumor, and expands a source of PD-1 in immune cells.
Collapse
Affiliation(s)
- Xiaodong Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China; Chinese Academy of Sciences (CAS) Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China; Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
| | - Xiaohui Yang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China; Chinese Academy of Sciences (CAS) Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China; Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
| | - Chang Huang
- Affiliated Hospital of ZunYi Medical University, Zunyi 563000, China
| | | | - Haojing Zang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yinmin Gu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
| | - Yibi Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China; Chinese Academy of Sciences (CAS) Key Laboratory of Biomedical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China; Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
| | | | - Chang Zhang
- Department of Oncology, The Key Laboratory of Advanced Interdisciplinary Studies, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510530, China
| | - Fang Guo
- Shanxi University, Taiyuan, 030001, China
| | - Songzhe Wu
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China
| | - Ao Ding
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rong Yin
- Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210096, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, 100850, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
16
|
Gupta A, Bhardwaj S, Ghorai S, Ahmed R, Agarwal S, Mukherjee G, Desai KV. Potential applications of gene expression profiles obtained from circulating extracellular vesicles in breast cancer. THE JOURNAL OF LIQUID BIOPSY 2025; 7:100287. [PMID: 40027231 PMCID: PMC11863812 DOI: 10.1016/j.jlb.2025.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Background Liquid biopsy-based biomarkers offer several advantages since they are minimally invasive, can be useful in longitudinal monitoring of the disease and have higher patient compliance. We describe a protocol using minimal volumes of archival and prospective serum/plasma samples to define the RNA contents of EVs and discuss its benefits and limitations. Methods RNA-seq analysis of matched tumor biopsy, circulating EVs from breast cancer patients (EV-C, n = 26) and healthy donors (EV-H, n = 4) was performed and differentially expressed genes were validated by RT-PCR in a separate series of samples (EV-C, n = 32 and EV-H, n = 22). A total of 84 samples were studied. Results RNA-seq data from 500 μl serum samples yielded more than 17000 genes, of which 320 were DEGs (adjusted p value ≤ 0.05) between EV-C and EV-H samples. Pathways for Myc V1, reactive oxygen species, angiogenesis, allograft rejection and Interferon regulated genes were over-represented in EV-C samples. Computational deconvolution algorithms for cell signatures identified immune cells such as Th1 and memory T-cells, endothelial cells, and osteoblasts from the stromal compartment as significant. Top 6 genes were validated by qRT-PCR in all samples (n = 84) and they consistently and correctly classified cancer and healthy groups. An independent set of 374 and 640 DEGs could segregate ER positive/ER negative and non-metastatic versus metastatic samples, respectively. EVs from metastatic samples had higher variability in gene expression patterns whereas those from non-metastatic samples showed a better correlation. Conclusion By using low serum amounts successfully for EV transcriptomics, we demonstrate that a minimally invasive technique could be converted to a microinvasive format. These data lay the foundation for EV RNA based biomarker discovery for segregating breast cancers.
Collapse
Affiliation(s)
- Aritra Gupta
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, PhD Program, India
| | - Siddharth Bhardwaj
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Sayan Ghorai
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, PhD Program, India
| | - Rosina Ahmed
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Sanjit Agarwal
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Geetashree Mukherjee
- Tata Medical Centre, 14 MAR (DH Block), New Town, Rajarhat, Kolkata, 700160, India
| | - Kartiki V. Desai
- Biotechnology Research Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| |
Collapse
|
17
|
Wang Y, Yi H, Huang K, Zeng Y, Miao P, Zhang Y, Hu N. 2-Mercaptoethanol enhances the yield of exosomes showing therapeutic potency in alleviating spinal cord injury mice. Life Sci 2025; 364:123451. [PMID: 39923836 DOI: 10.1016/j.lfs.2025.123451] [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: 12/11/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Limited passage numbers of mesenchymal stem cells (MSCs) present challenges in producing sufficient exosomes for spinal cord injury (SCI) treatment. OBJECTIVES This study investigates whether β-mercaptoethanol (BME) preconditioning of MSCs can increase exosome yield for SCI therapy. METHODS Exosomal content was analyzed using silver staining and SYBR Gold staining. Cell viability was assessed via CCK-8 and EdU assays. IL-1β, IL-6, TNF-α, and MCP-1 levels were measured by enzyme-linked immunosorbent assay (ELISA). Neuronal differentiation influenced by astrocytes was evaluated through neurite outgrowth and migration assays. Neuronal survival and motor function recovery in SCI mice were assessed using TUNEL staining, the Basso Mouse Scale (BMS), muscle strength tests, and motor evoked potential (MEP) measurements. RESULTS BME treatment significantly increased exosome quantity, including proteins and microRNAs, without drastic changes in exosomal content spectrum. Exosomes from BME-treated MSCs more effectively suppressed IL-1β, IL-6, TNF-α, and MCP-1 secretion by astrocytes, reducing neuronal inflammation. Yap1 activation reduced the exosomes' inhibitory effects on inflammatory cytokines. Mice treated with exosomes from BME-treated MSCs showed better outcomes: lower GFAP and C3 expression, reduced inflammation, increased NF-H levels, higher BMS scores, and greater MEP peaks. Exosome treatment also reduced bladder volume, residual urine, and the time to regain spontaneous urination after uroschesis. CONCLUSION BME preconditioning enhances exosome yield from hUC-MSCs, offering improved therapeutic potential for SCI.
Collapse
Affiliation(s)
- Yang Wang
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China.
| | - Hanxiao Yi
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Yuexiu District, Guangzhou, Guangdong Province, China.
| | - Keqi Huang
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China
| | - Yi Zeng
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China
| | - Ping Miao
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China.
| | - Yating Zhang
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China
| | - Nan Hu
- Department of Rehabilitation, The Second Affiliated Hospital, Guangzhou Medical University, Number 250 Changgang East Road, Haizhu District, Guangzhou, Guangdong Province, China.
| |
Collapse
|
18
|
Patel B, Gaikwad S, Prasad S. Exploring the significance of extracellular vesicles: Key players in advancing cancer and possible theranostic tools. CANCER PATHOGENESIS AND THERAPY 2025; 3:109-119. [PMID: 40182121 PMCID: PMC11963151 DOI: 10.1016/j.cpt.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 04/05/2025]
Abstract
Metastasis remains a critical challenge in cancer treatment and the leading cause of cancer-related mortality. Ongoing research has demonstrated the key role of extracellular vesicles (EVs) in facilitating communication between distant organs. Cancer cells release a substantial number of EVs that carry distinct cargo molecules, including oncogenic proteins, DNA fragments, and various RNA species. Upon uptake, these cargo molecules profoundly influence the biology of both normal and cancerous cells. This review consolidates the understanding of how EVs promote tumorigenesis by regulating processes such as proliferation, migration, metastasis, angiogenesis, stemness, and immunity. The exploration of EVs as a non-invasive method for cancer detection holds great promise, given that different cancer types exhibit unique protein and RNA signatures that can serve as valuable biomarkers for early diagnosis. Furthermore, growing interest exists in the potential bioengineering EVs for use as prospective therapeutic tools for cancer treatment.
Collapse
Affiliation(s)
- Bhaumik Patel
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutic and Biotechnology, Texas Tech University Health Science Center, Abilene, TX 79601, USA
| | - Sahdeo Prasad
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
19
|
Xiong Y, Lu X, Li B, Xu S, Fu B, Sha Z, Tian R, Yao R, Li Q, Yan J, Guo D, Cong Z, Du Y, Lin X, Wu H. Bacteroides Fragilis Transplantation Reverses Reproductive Senescence by Transporting Extracellular Vesicles Through the Gut-Ovary Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409740. [PMID: 39805029 PMCID: PMC11884595 DOI: 10.1002/advs.202409740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The diverse and dynamic population of microorganisms present in the gut microbiota may affect host health. There are evidences to support the role of gut microbiota as a key player in reproductive development. Unfortunately, the relationship between reproductive disorders caused by aging and gut microbiota remains largely unknown. Here, it is shown for the first time that gut microorganism Bacteroides fragilis (BF) transplantation ameliorates ovarian aging by transporting extracellular vesicles (EVs) through the gut-ovary axis. Mechanistically, miR-1246 is enriched in EVs derived from BF-treated intestinal cells, and these miR-1246-enriched EVs are transferred to ovaries, thereby effectively improving reproductive senescence by reducing oxidative stress in the ovaries. Specifically, miR-1246 reduces the ubiquitination of p62 and stabilizes the protein level of p62 by targeting E3 ligase SKP2. Then Keap1-Nrf2 complex is dissociated and Keap1 is recruited to form the p62-Keap1 complex. With the dissociation of Keap1-Nrf2 complex, Nrf2 is released and activated, thus promoting the transcription of antioxidant enzymes and relieving reproductive senescence. Collectively, the data indicates that intestinal cell-derived EVs serve as natural information carriers in the crosstalk between the gut and the ovary, and intestinal microorganism transplantation is a promising approach for the treatment of ovarian dysfunction diseases.
Collapse
Affiliation(s)
- Yan Xiong
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Xiaoxue Lu
- Department of Clinical Microbiology and ImmunologyCollege of Pharmacy and Medical LaboratoryArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Bohao Li
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Shiyao Xu
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Beibei Fu
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Zhou Sha
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Rong Tian
- Department of pathologyChongqing Hygeia HospitalChongqing401331China
| | - Rui Yao
- Department of pathologyChongqing Hygeia HospitalChongqing401331China
| | - Qian Li
- Department of Clinical Microbiology and ImmunologyCollege of Pharmacy and Medical LaboratoryArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Jingmin Yan
- Department of Clinical Microbiology and ImmunologyCollege of Pharmacy and Medical LaboratoryArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Dong Guo
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Zixuan Cong
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Yongliang Du
- School of Life SciencesChongqing UniversityChongqing401331China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and ImmunologyCollege of Pharmacy and Medical LaboratoryArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Haibo Wu
- School of Life SciencesChongqing UniversityChongqing401331China
| |
Collapse
|
20
|
Sun J, Luo J, Liu J, Wu H, Li Y, Xu Y, Liu L, Liu X, Zhang Q. Cancer-secreted exosomal miR-1825 induces angiogenesis to promote colorectal cancer metastasis. Cancer Cell Int 2025; 25:63. [PMID: 39987450 PMCID: PMC11847347 DOI: 10.1186/s12935-025-03674-5] [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: 10/12/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Angiogenesis is one of the important factors related to tumorigenesis, invasion, and metastasis. Cancer-secreted exosomes are essential mediators of intercellular cross-talk and participate in angiogenesis and metastasis. Unveiling the mechanism of angiogenesis is an important way to develop anti-angiogenesis therapeutic strategies to against cancer progression. METHODS miR-1825 expression and relationship with microvascular density were validated in colorectal cancer (CRC) by in situ hybridization (ISH) staining and immunohistochemistry (IHC). Sequential differential centrifugation, transmission electron microscopy, and western blotting analysis were used to extract and characterize exosomes. The effort of exosomal miR-1825 on endothelial cells was examined by transwell assay, wound healing assay, tube formation assay, and aortic ring assay. The relationship of miR-1825, ING1 and the downstream pathway were analyzed by western blot, RT-PCR, Immunofluorescence, and dual-luciferase reporter system analysis. RESULTS Exosomal miR-1825 is associated with angiogenesis in CRC and is enriched in exosomes extracted from the serum of CRC patients. The CRC-secreted exosomal miR-1825 can be transferred into vascular endothelial cells, promoting endothelial cell migration and tube formation in vitro, and facilitating angiogenesis and tumor metastasis in vivo. Mechanistically, exosomal miR-1825 regulates angiogenesis and tumor metastasis by suppressing inhibitor of growth family member 1 (ING1) and activating the TGF-β/Smad2/Smad3 signaling pathway in the recipient HUVECs. CONCLUSIONS Our study demonstrated the CRC-secreted exosomal miR-1825 could be transferred to vascular endothelial cells, subsequently leads to the inhibition of ING1 and the activation of the TGF-β/Smad2/Smad3 signaling pathway, thereby promoting angiogenesis and liver metastasis in CRC. Exosomal miR-1825 is thus a potential diagnostic and therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Jingbo Sun
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Junjie Luo
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Jialong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yanyan Li
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yangwei Xu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Xiaolong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, 183 West Zhongshan Avenue, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
21
|
Xiao N, Li Q, Liang G, Qian Z, Lin Y, Zhang H, Fu Y, Yang X, Zhang CT, Yang J, Liu A. Regulatory Roles of Exosomes in Aging and Aging-Related Diseases. Biogerontology 2025; 26:61. [PMID: 39966192 DOI: 10.1007/s10522-025-10200-7] [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: 11/09/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
Exosomes are small vesicles with diameters ranging from 30 to 150 nm. They originate from cellular endocytic systems. These vesicles contain a rich payload of biomolecules, including proteins, nucleic acids, lipids, and metabolic products. Exosomes mediate intercellular communication and are key regulators of a diverse array of biological processes, such as oxidative stress and chronic inflammation. Furthermore, exosomes have been implicated in the pathogenesis of infectious diseases, autoimmune disorders, and cancer. Aging is closely associated with the onset and progression of numerous diseases and is significantly influenced by exosomes. Recent studies have consistently highlighted the important functions of exosomes in the regulation of cellular senescence. Additionally, research has explored their potential to delay aging, such as the alleviatory effects of stem cell-derived exosomes on the aging process, which offers broad potential for the development and application of exosomes as anti-aging therapeutic strategies. This review aims to comprehensively investigate the multifaceted impact of exosomes while concurrently evaluating their potential applications and underscoring their strategic significance in advancing anti-aging strategies.
Collapse
Affiliation(s)
- Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yangguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Cun-Tai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, People's Republic of China.
| |
Collapse
|
22
|
Chen H, Liu L, Xing G, Zhang D, A. N, Huang J, Li Y, Zhao G, Liu M. Exosome tropism and various pathways in lung cancer metastasis. Front Immunol 2025; 16:1517495. [PMID: 40028322 PMCID: PMC11868168 DOI: 10.3389/fimmu.2025.1517495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer, characterized by its high morbidity and mortality rates, has the capability to metastasize to various organs, thereby amplifying its detrimental impact and fatality. The metastasis of lung cancer is a complex biological phenomenon involving numerous physiological transformations. Exosomes, small membranous vesicles enriched with biologically active components, are pivotal in mediating intercellular communication and regulating physiological functions due to their specificity and stability. Extensive research has elucidated the production and functions of exosomes in cancer contexts. Multitude of evidence demonstrates a strong association between lung cancer metastasis and exosomes. Additionally, the concept of the pre-metastatic niche is crucial in the metastatic process facilitated by exosomes. This review emphasizes the role of exosomes in mediating lung cancer metastasis and their impact on the disease's development and the progression to other tissues. Furthermore, it explores the potential of exosomes as biomarkers for lung cancer metastasis, offering significant insights for future clinical advancements.
Collapse
Affiliation(s)
- Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Drug Dispensing, The Third Hospital of Mianyang, Sichuan Mental Health Center, MianYang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Niumuqie A.
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianlin Huang
- Department of Pharmacy, Luzhou Naxi District People’s Hospital, Luzhou, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ge Zhao
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
23
|
Saadh MJ, Allela OQB, Kareem RA, Ballal S, Chahar M, Saini S, Prasad GVS, Sameer HN, Hamad AK, Athab ZH, Adil M. The role of exosomal non-coding RNAs in the breast cancer tumor microenvironment. Funct Integr Genomics 2025; 25:32. [PMID: 39891771 DOI: 10.1007/s10142-025-01531-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
The leading form of cancer affecting females globally is breast cancer, characterized by an unregulated growth of cells within the breast. Therefore, examining breast tissue is crucial in accurately identifying and treating this disease. Exosomes are very small enclosures bounded by a layer of cells and produced by a variety of cells present in the cancerous tissue surroundings. They play a crucial role in several biological functions in cancerous tumors. These exosomes carry non-coding RNAs (ncRNAs) and are discharged into the TME, where they are instrumental in the development and advancement of tumors. Additionally, the ncRNAs enclosed in exosomes act as significant mediators of communication within cells. Consequently, there is limited comprehension regarding the precise roles and targets of exosomal RNA in regulation, as research in this area is still in its preliminary phases. This piece provides a comprehensive overview of the latest studies on exosomes, delving into their impact on the behavior of cancer cells and immune cells. Moreover, it presents a compilation of the diverse forms of non-coding RNA molecules found in exosomes released by both cancerous and supportive cells, including circular RNAs, microRNAs, and long non-coding RNAs. Current research has proven the noteworthy influence that non-coding RNA molecules have on the progression, proliferation, drug resistance, and immune responses of breast cancer cells.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, 11831, Amman, Jordan
| | | | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Suman Saini
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, 140307, Mohali, Punjab, India
| | - G V Siva Prasad
- Department of Basic Sciences and Humanities, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, 64001, Dhi Qar, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, 00964, Baghdad, Iraq
| |
Collapse
|
24
|
Song Y, Kong H, Oh S, Kim SB. Plant-derived extracellular vesicles as nanocarriers for combination therapy enhancing paclitaxel-based regimens in breast cancer. BMB Rep 2025; 58:53-63. [PMID: 39978779 PMCID: PMC11875746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Breast cancer remains a leading cause of morbidity and mortality worldwide. Triple-negative breast cancer (TNBC) presents unique challenges owing to its aggressiveness and limited treatment options. Paclitaxel-based chemotherapy is widely used in breast cancer treatment. However, its efficacy is often limited by toxicity, multidrug resistance, and lack of targeted delivery. In response to these challenges, recent studies have focused on the use of extracellular vesicles (EVs), particularly plant-derived EVs, as innovative drug delivery systems capable of enhancing therapeutic outcomes and reducing adverse effects. Plant-derived EVs offer significant advantages owing to their biocompatibility, low immunogenicity, and scalability. They provide a natural platform for delivering chemotherapeutics such as paclitaxel and doxorubicin directly to tumor cells. This review explores the therapeutic potential of plant-derived EVs in breast cancer treatment, focusing on TNBC by examining their ability to improve drug stability, bioavailability, and selective targeting of cancer cells. Key studies on EVs derived from plants such as grapefruit, ginger, and tea leaves have demonstrated their capacity to deliver chemotherapeutic agents effectively while mitigating common side effects associated with conventional delivery methods. Although the use of plantderived EVs is still in early stages of research, findings suggest that that these nanocarriers can serve as transformative tools in oncology, providing a versatile and efficient platform for precise cancer treatment. This review highlights current landscape of research on plant-derived EVs, their application in breast cancer therapy, and future directions required to translate these findings into clinical practice. [BMB Reports 2025; 58(2): 53-63].
Collapse
Affiliation(s)
- Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Hyunseok Kong
- Department of Animal Science, Sahmyook University, Seoul 01795, Korea
| | - Soohwan Oh
- College of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sang Bum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| |
Collapse
|
25
|
Wang C, Zhang Y, Wang J, Han Y, Wang Y, Sun M, Liang Y, Huang M, Yu Y, Hu H, Liu H, Han L. Single-Cell Isolation Chip Integrated with Multicolor Barcode Array for High-Throughput Single-Cell Exosome Profiling in Tissue Samples. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411259. [PMID: 39659120 DOI: 10.1002/adma.202411259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/28/2024] [Indexed: 12/12/2024]
Abstract
Exosomes, functional biomarkers involved in cancer progression, have gained widespread attention for promoting tumor formation, growth, and metastasis. Current bulk exosome detections in bodily fluids enable cancer functional analysis, but average secretion levels from cell populations, losing parent cell information and ignoring exosome heterogeneity from diverse cell subgroups, necessitating an effective platform for analyzing single-cell exosome functional heterogeneity. Here, a high-throughput platform is presented, capable of efficient single-cell isolation and multi-color exosome phenotype analysis, as well as quantifying trace exosomes secreted by single cells. Photothermal-driven single-cell chips achieve significant single-cell isolation efficiency (≈97%) within 5 min, facilitating the ultra-high throughput single-cell exosome analysis. By conducting mass spectrometry and protein interaction of breast cancer exosome phenotypic proteins, key exosome phenotypes are identified. Tens of thousands of single cells from breast cancer cell lines, and clinical tissues are analyzed, revealing various subgroup differences. The study finds more CD44 and EGFR co-expressing exosome subgroups in breast cancer cell lines, while immune-evasion PD-L1 high-phenotype exosome subgroups are primarily presented in complex tumor microenvironments, especially in HER2-positive tissues. This platform offers powerful single-cell isolation, exosome quantification, and phenotypic analysis capabilities, making it a powerful tool for advancing single-cell exosome analysis in cancer research.
Collapse
Affiliation(s)
- Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
- Department of Integrated Circuits, Shandong University, Jinan, 250100, China
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, China
| | - Yunrui Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Mingyuan Sun
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yanbo Liang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Miao Huang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huili Hu
- School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
- Department of Integrated Circuits, Shandong University, Jinan, 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| |
Collapse
|
26
|
Sergi CM, Minervini F. DICER1: The Argonaute Endonuclease Family Member and Its Role in Pediatric and Youth Pathology. BIOLOGY 2025; 14:93. [PMID: 39857323 PMCID: PMC11761906 DOI: 10.3390/biology14010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
In 2001, two enzyme-encoding genes were recognized in the fruit fly Drosophila melanogaster. The genetic material, labeled Dicer-1 and Dicer-2, encodes ribonuclease-type enzymes with slightly diverse target substrates. The human orthologue is DICER1. It is a gene, which has been positioned on chromosome 14q32.13. It contains 27 exons, which are linking the two enzyme domains. DICER1 is found in all organ systems. It has been proved that it is paramount in human development. The protein determined by DICER1 is a ribonuclease (RNase). This RNase belongs to the RNase III superfamily, formally known as 'endoribonuclease'. It has been determined that the function of RNase III proteins is set to identify and degrade double-stranded molecules of RNA. DICER1 is a vital "housekeeping" gene. The multi-domain enzyme is key for small RNA processing. This enzyme functions in numerous pathways, including RNA interference paths, DNA damage renovation, and response to viruses. At the protein level, DICER is also involved in several human diseases, of which the pleuro-pulmonary blastoma is probably the most egregious entity. Numerous studies have determined the full range of DICER1 functions and the corresponding relationship to tumorigenic and non-neoplastic diseases. In fact, genetic mutations (somatic and germline) have been detected in DICER1 and are genetically associated with at least two clinical syndromes: DICER1 syndrome and GLOW syndrome. The ubiquity of this enzyme in the human body makes it an exquisite target for nanotechnology-supported therapies and repurposing drug approaches.
Collapse
Affiliation(s)
- Consolato M. Sergi
- Division of Anatomic Pathology, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Fabrizio Minervini
- Division of Thoracic Surgery, Cantonal Hospital Lucerne, 6000 Lucerne, Switzerland;
| |
Collapse
|
27
|
Li H, Lin H, Fan T, Huang L, Zhou L, Tian X, Zhao R, Zhang Y, Yang X, Wan L, Zhong H, Jiang N, Wei C, Chen W, Hou L. Exosomes derived from syncytia induced by SARS-2-S promote the proliferation and metastasis of hepatocellular carcinoma cells. Front Cell Infect Microbiol 2025; 14:1415356. [PMID: 39844837 PMCID: PMC11750861 DOI: 10.3389/fcimb.2024.1415356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/29/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) is characterized by fever, fatigue, dry cough, dyspnea, mild pneumonia and acute lung injury (ALI), which can lead to acute respiratory distress syndrome (ARDS), and SARS-CoV-2 can accelerate tumor progression. However, the molecular mechanism for the increased mortality in cancer patients infected with COVID-19 is unclear. Methods Colony formation and wound healing assays were performed on Huh-7 cells cocultured with syncytia. Exosomes were purified from the cell supernatant and verified by nanoparticle tracking analysis (NTA), Western blot (WB) analysis and scanning electron microscopy (SEM). Differentially expressed proteins in syncytia-derived exosomes (Syn-Exos) and their functions was analyzed by Proteomic sequencing. Syn-Exo-mediated promotion of hepatocellular carcinoma cells was measured by CCK-8 and Transwell migration assays. The mechanism by which Syn-Exos promote tumor growth was analyzed by Western blotting. A patient-derived xenotransplantation (PDX) mouse model was constructed to evaluate the pathological role of the SARS-CoV-2 spike protein (SARS-2-S). The number of syncytia in the tumor tissue sections was determined by immunofluorescence analysis. Results Syncytium formation promoted the proliferation and migration of hepatocellular carcinoma cells. Proteomic sequencing revealed that proteins that regulate cell proliferation and metastasis in Syn-Exos were significantly upregulated. Syn-Exos promote the proliferation and migration of hepatocellular carcinoma cells. Animal experiments showed that a pseudotyped lentivirus bearing SARS-2-S (SARS-2-Spp) promoted tumor development in PDX mice. More syncytia were found in tumor tissue from SARS-2-Spp mice than from VSV-Gpp mice. Conclusions Syn-Exos induced by SARS-2-S can promote the proliferation and metastasis of hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Huilong Li
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Haotian Lin
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Tinghui Fan
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Linfei Huang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Li Zhou
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaoyu Tian
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ruzhou Zhao
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yanhong Zhang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Luming Wan
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Nan Jiang
- Department of Pharmacy, Medical Supplies Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Congwen Wei
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wei Chen
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
28
|
Can C, Yang X, Jia H, Wu H, Guo X, Wei Y, Jia Z, Liu W, Zhang A, He N, Zhang H, Ma D. Exosomal circ_0006896 promotes AML progression via interaction with HDAC1 and restriction of antitumor immunity. Mol Cancer 2025; 24:4. [PMID: 39762891 PMCID: PMC11702195 DOI: 10.1186/s12943-024-02203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Drug resistance and immune escape continue to contribute to poor prognosis in AML. Increasing evidence suggests that exosomes play a crucial role in AML immune microenvironment. METHODS Sanger sequencing, RNase R and fluorescence in situ hybridization were performed to confirm the existence of circ_0006896. The role of circ_0006896 in the progression of AML was assessed by in vitro and in vivo functional experiments. Flow cytometry, RT-qPCR and adoptive T cell-transfer immunotherapy were conducted to assess the function of exosomal circ_0006896 in CD8+ T cell dysfunction. RNA pull-down assay, mass spectrometry, immunofluorescence, co-immunoprecipitation and western blot were performed to identify and confirm the circ_0006896 interacting proteins. RESULTS CircRNA expression patterns in exosomes differ significantly between AML and controls compared to lncRNAs or mRNAs. A new crucial exosomal circRNA, circ_0006896, is upregulated in both AML cells and exosomes and correlates with the prognosis and relapse of AML. In vitro and in vivo studies suggest that circ_0006896 significantly promotes AML cell proliferation, reduces chemotherapy sensitivity, and more importantly, impairs the efficacy of adoptive T cell-transfer immunotherapy. Mechanistically, circ_0006896 physically interacts with the catalytic domain of histone deacetylase HDAC1, decreasing histone H3 acetylation, and impairing the transcription of genes involved in arachidonic acid metabolism, ultimately inhibiting lipid peroxidation and ferroptosis in AML cells. Exosomal circ_0006896 disrupts CD8+ T cell function by interacting with HDAC1, impairing LEF1 transcription and subsequently decreasing the expression of cytotoxic molecules IFN-γ and Granzyme B. CONCLUSIONS We demonstrate a self-driven progression mediated by exosomal circRNAs and CD8+ T cells, highlighting the potential of targeting circRNAs in AML immunotherapy.
Collapse
MESH Headings
- Histone Deacetylase 1/metabolism
- Histone Deacetylase 1/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Humans
- RNA, Circular/genetics
- Mice
- Exosomes/metabolism
- Animals
- Disease Progression
- Cell Proliferation
- Cell Line, Tumor
- Tumor Microenvironment/immunology
- Prognosis
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Biomarkers, Tumor
- Xenograft Model Antitumor Assays
- Female
- Male
- Disease Models, Animal
Collapse
Affiliation(s)
- Can Can
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Xinyu Yang
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Hexiao Jia
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Hanyang Wu
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaodong Guo
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Yihong Wei
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Ziting Jia
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Wancheng Liu
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Amin Zhang
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Na He
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Hailei Zhang
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital of Shandong University, No.117, West of Wenhua Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
29
|
Zhang X, Upputuri PK, Coughlan MF, Kemble DJ, Khan U, Zakharov YN, Zhang L, Perelman LT, Qiu L. Dual-angle light scattering spectroscopy for calibration-free measurements of scatterer suspensions. OPTICS LETTERS 2025; 50:33-36. [PMID: 39718846 PMCID: PMC11891018 DOI: 10.1364/ol.543614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/16/2024] [Indexed: 12/26/2024]
Abstract
It can be difficult to employ optical techniques for analyzing biological structures smaller than or comparable to the wavelength of light, such as extracellular vesicles or some types of bacteria. Biological light scattering spectroscopy (LSS), developed to address this problem, has been successfully used for characterizing tissue on cellular and subcellular scales. At the same time, calibration with a reference sample of known optical properties can complicate LSS measurements. In this Letter, we present dual-angle LSS (daLSS), which is designed for calibration-free measurements of scatterer suspensions. It employs measurements of a sample at two distinct angles, which then allows system effects to be removed entirely. Not only does daLSS simplify and speed up the measurement procedure, but it also makes spectra more reproducible, an important feature for diagnostic techniques. We validated the technique by accurately reconstructing the sizes of polystyrene microspheres with diameters less than 100 nm and then demonstrated that not only are the daLSS spectra of several common bacteria strains easily distinguishable but they are also highly reproducible.
Collapse
Affiliation(s)
- Xuejun Zhang
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Paul K Upputuri
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Mark F Coughlan
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - David J Kemble
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard University, Boston, Massachusetts 02215 USA
| | - Umar Khan
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Yuri N Zakharov
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Lei Zhang
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Lev T Perelman
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| | - Le Qiu
- Center for Advanced Biomedical Imaging and Photonics, Harvard University, Boston, Massachusetts 02215 USA
| |
Collapse
|
30
|
Khanmohammadi S, Masrour M, Fallahtafti P, Hasani F. MicroRNA as a Potential Diagnostic and Prognostic Biomarker in Diffuse Large B-Cell Lymphoma: A Systematic Review and Meta-Analysis. Cancer Rep (Hoboken) 2025; 8:e70070. [PMID: 39854617 PMCID: PMC11760998 DOI: 10.1002/cnr2.70070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/19/2024] [Accepted: 11/12/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Recently, microRNAs (miRNAs) have been applied as biomarkers for diffuse large B-cell lymphoma (DLBCL) patients. Early diagnosis and management of DLBCL can improve patient survival and prognosis. AIMS This systematic review and meta-analysis aimed to evaluate the diagnostic and prognostic accuracy of miRNA biomarkers in DLBCL patients. METHODS We used the keywords "diffuse large B-cell lymphoma" and "microRNA" to search databases for original publications until June 14, 2023. Specificity, sensitivity, and AUC were used to assess diagnostic accuracy, and the prognostic value was assessed using the overall survival (OS) and progression-free survival (PFS) hazard ratio (HR). A subgroup analysis was performed based on the sample type acquired to investigate the heterogeneity. RESULTS Thirteen diagnostic and 33 prognostic studies were included from 839 articles. The Reitsma bivariate model estimated a sensitivity of 0.788 (95% CI: 0.733-0.834, p < 0.001), a specificity of 0.727 (95% CI: 0.654-0.790, p < 0.001), and an AUC of 0.824 in. The pooled AUC was 0.7385 (95% CI: 0.6847-0.7923, p < 0.0001). The pooled OS and PFS HRs (> 1) were 2.2847 (95% CI: 1.7248-3.0263, p < 0.0001) and 2.4883 (95% CI: 1.7367-3.5650, p < 0.0001). The pooled OS and PFS HRs (< 1) were 0.4965 (95% CI: 0.3576-0.6894, p < 0.0001) and 2.4883 (95% CI: 1.7367-3.5650, p < 0.0001). MiR-155 diagnostic values had a sensitivity of 0.710 (p > 0.1) and a specificity of 0.725 (p < 0.05), with an AUC of 0.776. miR-21 diagnostic values had an AUC of 0.8468 (p < 0.0001) and OS HR of 2.8938. CONCLUSION MicroRNAs could serve as a powerful diagnostic and prognostic tool in DLBCL.
Collapse
Affiliation(s)
- Shaghayegh Khanmohammadi
- School of MedicineTehran University of Medical SciencesTehranIran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical CenterTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mahdi Masrour
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Parisa Fallahtafti
- School of MedicineTehran University of Medical SciencesTehranIran
- Tehran Heart Center, Cardiovascular Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Fatemeh Hasani
- Golestan Research Center of Gastroenterology and HepatologyGolestan University of Medical SciencesGorganIran
| |
Collapse
|
31
|
Namdari M, McDonnell FS. Extracellular vesicles as emerging players in glaucoma: Mechanisms, biomarkers, and therapeutic targets. Vision Res 2025; 226:108522. [PMID: 39581065 PMCID: PMC11640964 DOI: 10.1016/j.visres.2024.108522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
In recent years, extracellular vesicles (EVs) have attracted significant scientific interest due to their widespread distribution, their potential as disease biomarkers, and their promising applications in therapy. Encapsulated by lipid bilayers these nanovesicles include small extracellular vesicles (sEV) (30-150 nm), microvesicles (100-1000 nm), and apoptotic bodies (100-5000 nm) and are essential for cellular communication, immune responses, biomolecular transport, and physiological regulation. As they reflect the condition and functionality of their originating cells, EVs play critical roles in numerous physiological processes and diseases. Therefore, EVs offer valuable opportunities for uncovering disease mechanisms, enhancing drug delivery systems, and identifying novel biomarkers. In the context of glaucoma, a leading cause of irreversible blindness, the specific roles of EVs are still largely unexplored. This review examines the emerging role of EVs in the pathogenesis of glaucoma, with a focus on their potential as diagnostic biomarkers and therapeutic agents. Through a thorough analysis of current literature, we summarize key advancements in EV research and identify areas where further investigation is needed to fully understand their function in glaucoma.
Collapse
Affiliation(s)
- Maral Namdari
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Pharmacology and Toxicology, University of Utah Salt Lake City, UT, USA.
| |
Collapse
|
32
|
Huang C, Li J, Xie Z, Hu X, Huang Y. Relationship between exosomes and cancer: formation, diagnosis, and treatment. Int J Biol Sci 2025; 21:40-62. [PMID: 39744442 PMCID: PMC11667803 DOI: 10.7150/ijbs.95763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/02/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes are a member of extracellular vesicles. However, their biological characteristics differ from those of other vesicles, and recently, their powerful functions as information molecules, biomarkers, and carriers have been demonstrated. Malignancies are the leading cause of high morbidity and mortality worldwide. The cure rate of malignancies can be improved by improving early screening rates and therapy. Moreover, a close correlation between exosomes and malignancies has been observed. An in-depth study of exosomes can provide new methods for diagnosing and treating tumors. Therefore, this study aimed to review, sort, and summarize such achievements, and present ideas and opinions on the application of exosomes in tumor treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajin Li
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zichuan Xie
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiangjun Hu
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, China
- Research Laboratory for Prediction and Evaluation of Chronic Diseases in the Elderly, National Clinical Research Center for Geriatric Diseases, China
- General Practice Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Amolegbe SM, Johnston NC, Ambrosi A, Ganguly A, Howcroft TK, Kuo LS, Labosky PA, Rudnicki DD, Satterlee JS, Tagle DA, Happel C. Extracellular RNA communication: A decade of NIH common fund support illuminates exRNA biology. J Extracell Vesicles 2025; 14:e70016. [PMID: 39815775 PMCID: PMC11735951 DOI: 10.1002/jev2.70016] [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: 08/03/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025] Open
Abstract
The discovery that extracellular RNAs (exRNA) can act as endocrine signalling molecules established a novel paradigm in intercellular communication. ExRNAs can be transported, both locally and systemically in virtually all body fluids. In association with an array of carrier vehicles of varying complexity, exRNA can alter target cell phenotype. This highlights the important role secreted exRNAs have in regulating human health and disease. The NIH Common Fund exRNA Communication program was established in 2012 to accelerate and catalyze progress in the exRNA biology field. The program addressed both exRNA and exRNA carriers, and served to generate foundational knowledge for the field from basic exRNA biology to future potential clinical applications as biomarkers and therapeutics. To address scientific challenges, the exRNA Communication program developed novel tools and technologies to isolate exRNA carriers and analyze their cargo. Here, we discuss the outcomes of the NIH Common Fund exRNA Communication program, as well as the evolution of exRNA as a scientific field through the analysis of scientific publications and NIH funding. ExRNA and associated carriers have potential clinical use as biomarkers, diagnostics, and therapeutics. Recent translational applications include exRNA-related technologies repurposed as novel diagnostics in response to the COVID-19 pandemic, the clinical use of extracellular vesicle-based biomarker assays, and exRNA carriers as drug delivery platforms. This comprehensive landscape analysis illustrates how discoveries and innovations in exRNA biology are being translated both into the commercial market and the clinic. Analysis of program outcomes and NIH funding trends demonstrate the impact of this NIH Common Fund program.
Collapse
Affiliation(s)
- Sara M. Amolegbe
- Office of the DirectorNational Institutes of HealthBethesdaMarylandUSA
| | - Nicolas C. Johnston
- National Institute on Drug AbuseNational Institutes of HealthBethesdaMarylandUSA
| | - Angela Ambrosi
- Office of the DirectorNational Institutes of HealthBethesdaMarylandUSA
| | - Aniruddha Ganguly
- National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - T. Kevin Howcroft
- National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Lillian S. Kuo
- National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | | | - Dobrila D. Rudnicki
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMarylandUSA
| | - John S. Satterlee
- National Institute on Drug AbuseNational Institutes of HealthBethesdaMarylandUSA
| | - Danilo A. Tagle
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMarylandUSA
| | - Christine Happel
- National Center for Advancing Translational SciencesNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
34
|
Linares-Rodríguez M, Blancas I, Rodríguez-Serrano F. The Predictive Value of Blood-Derived Exosomal miRNAs as Biomarkers in Breast Cancer: A Systematic Review. Clin Breast Cancer 2025; 25:e48-e55.e15. [PMID: 39054208 DOI: 10.1016/j.clbc.2024.06.016] [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: 12/21/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024]
Abstract
Breast cancer (BC) remains a widespread disease worldwide, despite advances in its detection and treatment. microRNAs (miRNAs) play a significant role in cancer, and their presence within exosomes may confer several advantages in terms of tumor initiation, propagation, immune evasion, and drug resistance compared to freely circulating miRNAs in the blood. The objective of this study was to conduct a systematic review to analyze the role of exosomal miRNAs present in serum or plasma as biomarkers in BC. Bibliographic sources were collected from various databases with no starting date limit until March 2023. The search terms used were related to "breast cancer," "microRNAs," and "exosomes." Following the search, inclusion and exclusion criteria were applied, resulting in a total of 46 articles. Data were extracted from the selected studies and summarized to indicate the miRNAs, type of dysregulation, sample source, number of patients and controls, and clinical relevance of the miRNAs. We carried out an enrichment study of the microRNAs that appeared in at least 3 studies, those that were suitable for selection were miR-16, miR-21 and miR-155. Exosomal miRNAs isolated from blood samples of patients diagnosed with BC could be valuable in the clinical setting. They could provide information about early diagnosis, disease progression, recurrence, treatment response, and metastases. It is crucial to reach a consensus on the specific exosomal miRNAs to detect and the most appropriate type of sample for comprehensive utilization of miRNAs as biomarkers for BC.
Collapse
Affiliation(s)
- Marina Linares-Rodríguez
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
| | - Isabel Blancas
- Department of Medicine, School of Medicine, University of Granada, Granada, Spain; Department of Medical Oncology, San Cecilio University Hospital, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.
| | - Fernando Rodríguez-Serrano
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.
| |
Collapse
|
35
|
Huang H, Wang Y, Gan J, Bu H, Li C, Li Y, Yang Y, Liu D. FRK exerts oncogenic effects by targeting NQO2 via exosomal miR-9-3p to regulate mitochondrial function. FASEB J 2024; 38:e70221. [PMID: 39625368 DOI: 10.1096/fj.202400951r] [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: 04/26/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 03/17/2025]
Abstract
The role of Fyn-related kinase (FRK) in promoting tumor progression and metabolism in non-small cell lung cancer (NSCLC) has been demonstrated in our earlier study, and here we further explored whether exosome could serve as a key player in the relevant regulatory mechanism. Exosomes were isolated from the culture medium of FRK-knockout (KO) cells and co-cultured with NSCLC cells, showing negative effects on cell proliferation, metastasis, metabolism, and mitochondrial function. Exosomal miR-9-3p was selected as an intermediate messenger through microRNA (miRNA) sequencing. The miR-9-3p inhibitor and exosome inhibitor GW4869 were then applied in the co-culture system to verify the contribution of exosomal miR-9-3p to cell malignant phenotype and mitochondrial function. Moreover, NQO2, a potential target of exosomal miR-9-3p, was also suggested to be involved in the regulation of mitochondrial functional status. In brief, this study revealed a novel molecular mechanism by which FRK promotes the malignant progression of NSCLC by targeting NQO2 via exosomal miR-9-3p to strengthen mitochondrial function.
Collapse
Affiliation(s)
- Hong Huang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Wang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiadi Gan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Bu
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cong Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongfeng Yang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Strum S, Evdokimova V, Radvanyi L, Spreafico A. Extracellular Vesicles and Their Applications in Tumor Diagnostics and Immunotherapy. Cells 2024; 13:2031. [PMID: 39682778 PMCID: PMC11639792 DOI: 10.3390/cells13232031] [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: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that have attracted significant attention in the investigation of human health and disease, including cancer biology and its clinical management. Concerning cancer, EVs have been shown to influence numerous aspects of oncogenesis, including tumor proliferation and metastasis. EVs can augment the immune system and have been implicated in virtually all aspects of innate and adaptive immunity. With immunotherapy changing the landscape of cancer treatment across multiple disease sites, it is paramount to understand their mechanisms of action and to further improve upon their efficacy. Despite a rapidly growing body of evidence supporting of the utility of EVs in cancer diagnostics and therapeutics, their application in clinical trials involving solid tumors and immunotherapy remains limited. To date, relatively few trials are known to incorporate EVs in this context, mainly employing them as biomarkers. To help address this gap, this review summarizes known applications of EVs in clinical trials and provides a brief overview of the roles that EVs play in cancer biology, immunology, and their proposed implications in immunotherapy. The impetus to leverage EVs in future clinical trials and correlative studies is crucial, as they are ideally positioned to synergize with advancements in multi-omics research to further therapeutic discovery and our understanding of cancer biology.
Collapse
Affiliation(s)
- Scott Strum
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | | | - Laszlo Radvanyi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
37
|
Shkurnikov M, Averinskaya D, Stekolshchikova E, Serkina A, Razumovskaya A, Silkina M, Antipenko I, Makarova J, Evtushenko E, Nikulin S, Tonevitsky A. IGFBP6 regulates extracellular vesicles formation via cholesterol abundance in MDA-MB-231 cells. Biochimie 2024; 227:77-85. [PMID: 38942135 DOI: 10.1016/j.biochi.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Breast cancer recurrence is associated with the growth of disseminated cancer cells that separate from the primary tumor before surgical treatment and hormonal therapy and form a metastatic niche in distant organs. We previously demonstrated that IGFBP6 expression is associated with the risk of early relapse of luminal breast cancer. Knockdown of IGFBP6 in MDA-MB-231 breast cancer cells increased their invasiveness, proliferation, and metastatic potential. In addition, the knockdown of IGFBP6 leads to impaired lipid metabolism. In this study, we demonstrated that the knockdown of the IGFBP6 gene, a highly selective inhibitor of IGF-II, led to a significant decline in the number of secreted extracellular vesicles (EVs) and altered cholesterol metabolism in MDA-MB-231 cells. Knockdown of IGFBP6 led to a decrease in the essential proteins responsible for the biogenesis of cholesterol LDLR and LSS, which reduced the amount by more than 13 times. In addition, the knockdown of IGFBP6 led to a possible change in the profile of adhesion molecules on the surface of EVs. The expression of L1CAM, IGSF3, EpCAM, CD24, and CD44 decreased, and the expression of EGFR increased. We can conclude that the negative prognostic value of low expression of this gene could be associated with increased activity of IGF2 in tumor-associated fibroblasts due to low secretion of IGFBP6 by tumor cells. In addition, changing the profile of adhesion molecules on the surface of tumor EVs may contribute to the more efficient formation of metastatic niches.
Collapse
Affiliation(s)
- Maxim Shkurnikov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia.
| | - Darya Averinskaya
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Elena Stekolshchikova
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anna Serkina
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Alexandra Razumovskaya
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia; P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre of the Ministry of Health of Russian Federation, Moscow, Russia
| | - Maria Silkina
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia; P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre of the Ministry of Health of Russian Federation, Moscow, Russia
| | - Ivan Antipenko
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Julia Makarova
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | | | - Sergey Nikulin
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia; P. Hertsen Moscow Oncology Research Institute-Branch of the National Medical Research Radiological Centre of the Ministry of Health of Russian Federation, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
38
|
Ji M, Li L, Yu J, Wu Z, Sheng Y, Wang F. New insights into the function and therapeutic potential of RNA-binding protein TRBP in viral infection, chronic metabolic diseases, brain disorders and cancer. Life Sci 2024; 358:123159. [PMID: 39447729 DOI: 10.1016/j.lfs.2024.123159] [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: 08/20/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
RNA-binding proteins (RBPs) and non-coding RNAs are crucial trans-acting factors that bind to specific cis-acting elements in mRNAs, thereby regulating their stability and translation. The trans-activation response (TAR) RNA-binding protein (TRBP) recognizes precursor microRNAs (pre-miRNAs), modulates miRNA maturation, and influences miRNA interference (mi-RNAi) mediated by the RNA-induced silencing complex (RISC). TRBP also directly binds and mediates the degradation of certain mRNAs. Thus, TRBP acts as a hub for regulating gene expression and influences a variety of biological processes, including immune evasion, metabolic abnormalities, stress response, angiogenesis, hypoxia, and metastasis. Aberrant TRBP expression has been proven to be closely related to the initiation and progression of diseases, such as viral infection, chronic metabolic diseases, brain disorders, and cancer. This review summarizes the roles of TRBP in cancer and other diseases, the therapeutic potential of TRBP inhibition, and the current status of drug discovery on TRBP.
Collapse
Affiliation(s)
- Minghui Ji
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingyu Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jialing Yu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhao Wu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuwen Sheng
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.
| |
Collapse
|
39
|
Yan H, Cai X, Zhang J, Zhao H, Wu H, Zhang J, Xu L, Liu S, Zang Y, Fu S. Gastric cancer cell-derived exosomal miRNA-128-3p promotes angiogenesis by targeting SASH1. Front Oncol 2024; 14:1440996. [PMID: 39664191 PMCID: PMC11631727 DOI: 10.3389/fonc.2024.1440996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Exosomes, key components of the tumour microenvironment, can mediate intercellular communication through the delivery of various signalling molecules, including microribonucleic acids (miRNAs), and ultimately participate in regulating the process of tumour development. In this study, we aimed to investigate the reason and mechanism by which exosomal miRNAs derived from gastric cancer cells affect carcinogenesis and metastasis. Among these miRNAs, microRNA-128-3p (miR-128-3p) was highly expressed in serum exosomes isolated from gastric cancer patients, as confirmed by high-throughput sequencing and subsequent experiments. Coculture of gastric cancer-derived exosomes overexpressing miR-128-3p with human umbilical vein endothelial cells (HUVECs) significantly enhanced HUVEC proliferation, migratio n and angiogenesis. Bioinformatics analysis suggested SASH1 as the target gene of miR-128-3p. The dual luciferase assay and Western blot analysis results confirmed that miR-128-3p directly targeted SASH1 to inhibit its expression in HUVECs. Therefore, this study provides preliminary evidence that gastric cancer-derived exosomal miR-128-3p promotes tumour angiogenesis by targeting SASH1, reveals the potential diagnostic and therapeutic value of cancer-derived exosomal miR-128-3p, and provides new insights into the novel molecular mechanisms regulating metastasis. This study provides further information for understanding the role of gastric cancer-derived exosomal miR-128-3p in cancer progression and to discover new therapeutic targets.
Collapse
Affiliation(s)
- Hao Yan
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Xinyu Cai
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianna Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongpeng Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongwen Wu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Jingbo Zhang
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Lanzhi Xu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Shizheng Liu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shanshan Fu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
40
|
Bin Islam MK, Marcus RK. Isolation and quantification of human urinary exosomes using a Tween-20 elution solvent from polyester, capillary-channeled polymer fiber columns. Anal Chim Acta 2024; 1329:343242. [PMID: 39396305 PMCID: PMC11471952 DOI: 10.1016/j.aca.2024.343242] [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: 03/13/2024] [Revised: 08/08/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Exosomes, a subset of extracellular vesicles (EVs), are a type of membrane-secreted vesicle essential for intercellular communication. There is a great deal of interest in developing methods to isolate and quantify exosomes to study their role in intercellular processes and as potential therapeutic delivery systems. Polyester, capillary-channeled polymer fiber columns and spin-down tips are highly efficient, low-cost means of exosome isolation. As the methodology evolves, there remain questions as to the optimum elution solvent for specific end-uses of the recovered vesicles; fundamental biochemistry, clinical diagnostics, or therapeutic vectors. RESULTS While both acetonitrile and glycerol have been proven highly successful in terms of EV recoveries in the hydrophobic interaction chromatography workflow, many biological studies entail the use of the non-ionic detergent, Tween-20, as a working solvent. Here we evaluate the use of Tween-20 as the elution solvent for the recovery of exosomes. A novel 10-min, two-step gradient elution method, employing 0.1 % v/v Tween-20, efficiently isolated EVs at a concentration of ∼1011 EV mL-1 from a 100 μL urine injection. Integration of absorbance and multi-angle light scattering detectors in standard HPLC instrumentation enables a comprehensive single-injection determination of eluted exosome concentration and sizes. Transmission electron microscopy verifies the retention of the vesicular structure of the exosomes. The micro-bicinchoninic acid protein quantification assay confirmed high-purity isolations of exosomes (∼99 % removal of background proteins) SIGNIFICANCE: The effective use of Tween-20 as an elution solvent for exosome isolation/purification using capillary-channeled polymer fiber columns adds greater versatility to the portfolio of the approach. The proposed method holds promise for a wide range of fundamental biochemistry, clinical diagnostics, and therapeutic applications, marking a significant advancement in EV-based methodologies.
Collapse
Affiliation(s)
- Md Khalid Bin Islam
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC, 29634-0973, USA
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, SC, 29634-0973, USA.
| |
Collapse
|
41
|
Jaffet J, Singh V, Schrader S, Mertsch S. The Potential Role of Exosomes in Ocular Surface and Lacrimal Gland Regeneration. Curr Eye Res 2024:1-14. [PMID: 39508276 DOI: 10.1080/02713683.2024.2424265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/01/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Dry eye disease (DED), a multifactorial disease of the lacrimal system, manifests itself in patients with various symptoms such as itching, inflammation, discomfort and visual impairment. In its most severe forms, it results in the breakdown of the vital tissues of lacrimal functional unit and carries the risk of vision loss. Despite the frequency of occurrence of the disease, there are no effective curative treatment options available to date. Treatment using stem cells and its secreted factors could be a promising approach in the regeneration of damaged tissues of ocular surface. The treatment using secreted factors as well as extracellular vesicles has been demonstrated beneficial effects in various ocular surface diseases. This review provides insights on the usage of stem cell derived exosomes as a promising therapy against LG dysfunction induced ADDE for ocular surface repair. METHODS In order to gain an overview of the existing research in this field, literature search was carried out using the PubMed, Medline, Scopus and Web of Science databases. This review is based on 164 publications until June 2024 and the literature search was carried out using the key words "exosomes", "lacrimal gland regeneration", "exosomes in lacrimal dysfunction". RESULTS The literature and studies till date suggest that exosomes and other secreted factors from stem cells have demonstrated beneficial effects on damaged ocular tissues in various ocular surface diseases. Exosomal cargo plays a crucial role in regenerating tissues by promoting homeostasis in the lacrimal system, which is often compromised in severe cases of dry eye disease. Exosome therapy shows promise as a regenerative therapy, potentially addressing the lack of effective curative treatments available for patients with dry eye disease. CONCLUSION Stem cell-derived exosomes represent a promising, innovative approach as a new treatment option for ADDE. By targeting lacrimal gland dysfunction and enhancing ocular surface repair, exosome therapy offers potential for significant advances in dry eye disease management. Future research is needed to refine the application of this therapy, optimize delivery methods, and fully understand its long-term efficacy in restoring ocular health.
Collapse
Affiliation(s)
- Jilu Jaffet
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Vivek Singh
- LV Prasad Eye Institute, Centre for Ocular Regeneration, Hyderabad, Telangana, India
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, Carl von Ossietzky University, Oldenburg, Germany
| |
Collapse
|
42
|
Cui H, Zheng T, Qian N, Fu X, Li A, Xing S, Wang XF. Aptamer-Functionalized Magnetic Ti 3C 2 Based Nanoplatform for Simultaneous Enrichment and Detection of Exosomes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402434. [PMID: 38970554 DOI: 10.1002/smll.202402434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/11/2024] [Indexed: 07/08/2024]
Abstract
Exosomes are nanovesicles secreted by cells, which play a crucial role in various pathological processes. Exosomes have shown great promise as tumor biomarkers because of the abundant secretion during tumor formation. The development of a convenient, efficient, and cost-effective method for simultaneously enriching and detecting exosomes is of utmost importance for both basic research and clinical applications. In this study, an aptamer-functionalized magnetic Ti3C2 composite material (Fe3O4@Ti3C2@PEI@DSP@aptamer@FAM-ssDNA) is prepared for the simultaneous enrichment and detection of exosomes. CD63 aptamers are utilized to recognize and capture the exosomes, followed by magnetic separation. The exosomes are then released by cleaving the disulfide bonds of DSP. Compared to traditional methods, Fe3O4@Ti3C2@PEI@DSP@aptamer@FAM-ssDNA exhibited superior efficiency in enriching exosomes while preserving their structural and functional integrity. Detection of exosome concentration is achieved through the fluorescence quenching of Ti3C2 and the competitive binding between the exosomes and a fluorescently labeled probe. This method exhibited a low detection limit of 4.21 × 104 particles mL-1, a number that is comparable to the state-of-the-art method in the detection of exosomes. The present study demonstrates a method of simultaneous enrichment and detection of exosomes with a high sensitivity, accuracy, specificity, and cost-effectiveness providing significant potential for clinical research and diagnosis.
Collapse
Affiliation(s)
- Hongyuan Cui
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory of Physics and Technology for Advanced Batteries (Ministry of Education), College of Physics, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
| | - Tianfang Zheng
- Key Laboratory of Physics and Technology for Advanced Batteries (Ministry of Education), College of Physics, Jilin University, Changchun, 130012, P. R. China
| | - Nana Qian
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
| | - Xueqi Fu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
| | - Aijun Li
- Key Laboratory of Physics and Technology for Advanced Batteries (Ministry of Education), College of Physics, Jilin University, Changchun, 130012, P. R. China
| | - Shu Xing
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, P. R. China
| | - Xiao-Feng Wang
- Key Laboratory of Physics and Technology for Advanced Batteries (Ministry of Education), College of Physics, Jilin University, Changchun, 130012, P. R. China
| |
Collapse
|
43
|
Yasui T, Natsume A, Yanagida T, Nagashima K, Washio T, Ichikawa Y, Chattrairat K, Naganawa T, Iida M, Kitano Y, Aoki K, Mizunuma M, Shimada T, Takayama K, Ochiya T, Kawai T, Baba Y. Early Cancer Detection via Multi-microRNA Profiling of Urinary Exosomes Captured by Nanowires. Anal Chem 2024; 96:17145-17153. [PMID: 39422334 PMCID: PMC11525924 DOI: 10.1021/acs.analchem.4c02488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Multiple microRNAs encapsulated in extracellular vesicles (EVs) including exosomes, unique subtypes of EVs, differ in healthy and cancer groups of people, and they represent a warning sign for various cancer scenarios. Since all EVs in blood cannot be transferred from donor to recipient cells during a single blood circulation, kidney filtration could pass some untransferred EVs from blood to urine. Previously, we reported on the ability of zinc oxide nanowires to capture EVs based on surface charge and hydrogen bonding; these nanowires extracted massive numbers of microRNAs in urine, seeking cancer-related microRNAs through statistical analysis. Here, we report on the scalability of the nanowire performance capability to comprehensively capture EVs, including exosomes, in urine, extract microRNAs from the captured EVs in situ, and identify multiple microRNAs in the extracted microRNAs differing in noncancer and lung cancer subjects through machine learning-based analysis. The nanowire-based extraction allowed the presence of about 2500 species of urinary microRNAs to be confirmed, meaning that urine includes almost all human microRNA species. The machine learning-based analysis identified multiple microRNAs from the extracted microRNA species. The ensembles could classify cancer and noncancer subjects with the area under the receiver operating characteristic curve of 0.99, even though the former were staged early.
Collapse
Affiliation(s)
- Takao Yasui
- Department
of Life Science and Technology, Tokyo Institute
of Technology, Nagatsuta 4259, Midori-ku, Yokohama 226-8501, Japan
- Institute
of Quantum Life Science, National Institutes
for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Craif
Inc., 3-38-14-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Atsushi Natsume
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Craif
Inc., 3-38-14-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Kawamura
Medical Society, Gifu 501-3144, Japan
| | - Takeshi Yanagida
- Department
of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- The
Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Kazuki Nagashima
- Research
Institute for Electronic Science (RIES), Hokkaido University, N21W10, Kita, Sapporo, Hokkaido 001-0021, Japan
| | - Takashi Washio
- The
Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Yuki Ichikawa
- Craif
Inc., 3-38-14-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kunanon Chattrairat
- Department
of Life Science and Technology, Tokyo Institute
of Technology, Nagatsuta 4259, Midori-ku, Yokohama 226-8501, Japan
| | - Tsuyoshi Naganawa
- Department
of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Mikiko Iida
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Yotaro Kitano
- Department
of Neurosurgery, School of Medicine, Nagoya
University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kosuke Aoki
- Department
of Neurosurgery, School of Medicine, Nagoya
University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Mika Mizunuma
- Craif
Inc., 3-38-14-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Taisuke Shimada
- Institute
of Quantum Life Science, National Institutes
for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Kazuya Takayama
- Craif
Inc., 3-38-14-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takahiro Ochiya
- Department
of Molecular and Cellular Medicine, Tokyo
Medical University, 6-7-1 Nishishinjyuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Tomoji Kawai
- The
Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Yoshinobu Baba
- Institute
of Quantum Life Science, National Institutes
for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Institute
of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| |
Collapse
|
44
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 PMCID: PMC11799831 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
45
|
Dubrovsky G, Ross A, Jalali P, Lotze M. Liquid Biopsy in Pancreatic Ductal Adenocarcinoma: A Review of Methods and Applications. Int J Mol Sci 2024; 25:11013. [PMID: 39456796 PMCID: PMC11507494 DOI: 10.3390/ijms252011013] [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: 09/12/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a malignancy with one of the highest mortality rates. One limitation in the diagnosis and treatment of PDAC is the lack of an early and universal biomarker. Extensive research performed recently to develop new assays which could fit this role is available. In this review, we will discuss the current landscape of liquid biopsy in patients with PDAC. Specifically, we will review the various methods of liquid biopsy, focusing on circulating tumor DNA (ctDNA) and exosomes and future opportunities for improvement using artificial intelligence or machine learning to analyze results from a multi-omic approach. We will also consider applications which have been evaluated, including the utility of liquid biopsy for screening and staging patients at diagnosis as well as before and after surgery. We will also examine the potential for liquid biopsy to monitor patient treatment response in the setting of clinical trial development.
Collapse
Affiliation(s)
- Genia Dubrovsky
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.D.); (A.R.)
- Pittsburgh VA Medical Center, Pittsburgh, PA 15240, USA
| | - Alison Ross
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA; (G.D.); (A.R.)
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Michael Lotze
- Departments of Surgery, Immunology, and Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
46
|
Zhang C, Zhang L, Huang Q, Jiang S, Peng T, Wang S, Xu X. Diagnostic and screening potential of plasma exosome miR‑99b‑5p and its combination with other miRNAs for colorectal cancer. Oncol Lett 2024; 28:461. [PMID: 39119230 PMCID: PMC11307556 DOI: 10.3892/ol.2024.14594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/15/2024] [Indexed: 08/10/2024] Open
Abstract
Extracellular vesicles (EVs) secreted by tumor cells have been documented to hold viable biomarker potential. Therefore, the present study evaluated the potential clinical value of EV-microRNAs (miRNAs or miRs) in the plasma exosomes of patients with colorectal cancer (CRC) for the early diagnosis and screening of CRC. In total, 95 plasma samples were collected at The Third Affiliated Hospital of Guangzhou Medical University (Guangzhou, China) between 2017 and 2019. Specifically, 68 samples were from patients with CRC and 27 were from healthy control (HC) donors. High-throughput sequencing was used to detect the expression of miRNAs in the isolated plasma EVs, which was subsequently verified by reverse transcription-quantitative PCR. Receiver operating characteristic (ROC) curves were used to analyze the diagnostic potential of single and combined miRNAs for CRC. Bioinformatics analysis was employed to predict the target genes of candidate miRNAs. Compared with those in the HC group, the CRC group expressed higher levels of miR-99b-5p and miR-409-3p, especially during the early stages of CRC. Clinicopathological analysis confirmed the higher expression levels of miR-99b-5p during the early stages, as well as higher expression levels in the colon compared with those in the rectum. ROC curve analysis revealed that the area under the curve (AUC) of miR-99b-5p for the diagnosis of early CRC was 73.5% (P=0.007). The early diagnostic capability of miR-99b-5p combined with miR-409-3p for CRC was evaluated, and the AUC was found to be 74.1% (P=0.006). In addition, the AUC of the combination of miR-99b-5p, miR-409-3p and carcinoembryonic antigen was 81.2% (P<0.001), indicating that this three-parameter combination displayed higher diagnostic power compared with any single miRNA for early CRC screening. The results from the present study suggest that the expression of miR-99b-5p in plasma exosomes is significantly upregulated in CRC, which holds potential for the early diagnosis of this cancer type. Such potential can be enhanced further by combining it with other miRNAs. Therefore, the present study provides a comprehensive but preliminary insight for the viability of miR-99b-5p (alone or combined with other miRNAs) for CRC diagnosis, which requires further exploration in the future.
Collapse
Affiliation(s)
- Chang Zhang
- Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an, Shanxi 710032, P.R. China
- Department of Aviation Medicine, The First Affiliated Hospital, Air Force Medical University, Xi'an, Shanxi 710032, P.R. China
| | - Limei Zhang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Qiyuan Huang
- Nursing School, Guangzhou Medical University, Guangzhou, Guangdong 510030, P.R. China
| | - Siyuan Jiang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Tao Peng
- Sino-French Hoffmann Institute, Guangzhou Hoffman Institute of Immunology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Republic of Singapore
| | - Xuehu Xu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| |
Collapse
|
47
|
Bitiņa-Barlote Ē, Plonis J, Andrejeva M, Vjaters E, Gardovskis J, Daneberga Z, Miklaševičs E, Nakazawa-Miklaševiča M. Profiles of urinal exosomal miRNAs derived from bladder cancer. Cent European J Urol 2024; 77:361-374. [PMID: 40115475 PMCID: PMC11921953 DOI: 10.5173/ceju.2023.279.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 06/21/2024] [Indexed: 03/23/2025] Open
Abstract
Introduction Exosomes contain nucleic acids and proteins inside of them. These are suggested as cell-cell communication materials and it is considered that they can modulate the status of other cells. Material and methods To understand the bladder cancer (BC) related exosomal microRNAs (miRNAs), we compared the 752 urine exosomal miRNAs in healthy control (n = 7), low grade (LG) BC (n = 6) and high grade (HG) BC (n = 6) by RT-qPCR. Results The differential expressing (DE) urine exosomal miRNAs (2 > fold regulation) were 96 and 78 in LG and HG, respectively. Our exosomal miRNAs profiles cover many miRNAs which have been reported in BC patients' tissues and other biofluids. Most DE exosomal miRNAs were up-regulated in the profiles. Seven up-regulated exosomal miRNAs in the LG group (miR-28-5p, miR-16-5p, miR-28-3p, miR-24-3p, miR-25-3p, miR-19b-3p and miR10b-5p) and 3 miRNAs in the HG group (miR-150-5p, miR-28-5p and miR28-3p) were found as directly TP53 targeting. Twenty-two and 18 PTEN targeting miRNAs were observed in up-regulated miRNAs of LG and HG. The target genes of these exosomal miRNAs and their interaction network predicted that the TP53 is the strongest hub gene in both BC groups exosomal miRNA networks. Several DE miRNAs were found that could potentially be used as biomarkers for the diagnosis of BC. Conclusions Profiles of urinal exosomal miRNAs derived from BC manifested potentially epigenetic regulation of the TP53 and PTEN genes as compared to other oncogenes and tumour suppressors.
Collapse
Affiliation(s)
- Ērika Bitiņa-Barlote
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Juris Plonis
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Margarita Andrejeva
- Center of Oncology, Pauls Stradiņš Clinical University Hospital, Riga, Latvia
| | - Egils Vjaters
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Jānis Gardovskis
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
| | - Zanda Daneberga
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
| | - Edvīns Miklaševičs
- Department of Biology and Microbiology, Rīga Stradiņš University, Latvia
| | | |
Collapse
|
48
|
Li R, Ji Y, Ye R, Tang G, Wang W, Chen C, Yang Q. Potential therapies for non-coding RNAs in breast cancer. Front Oncol 2024; 14:1452666. [PMID: 39372872 PMCID: PMC11449682 DOI: 10.3389/fonc.2024.1452666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 10/08/2024] Open
Abstract
Breast cancer (BC) is one of the frequent tumors that seriously endanger the physical and mental well-being in women with strong heterogeneity, and its pathogenesis involves multiple risk factors. Depending on the type of BC, hormonal therapy, targeted therapy, and immunotherapy are the current systemic treatment options along with conventional chemotherapy. Despite significant progress in understanding BC pathogenesis and therapeutic options, there is still a need to identify new therapeutic targets and develop more effective treatments. According to recent sequencing and profiling studies, non-coding (nc) RNAs genes are deregulated in human cancers via deletion, amplification, abnormal epigenetic, or transcriptional regulation, and similarly, the expression of many ncRNAs is altered in breast cancer cell lines and tissues. The ability of single ncRNAs to regulate the expression of multiple downstream gene targets and related pathways provides a theoretical basis for studying them for cancer therapeutic drug development and targeted delivery. Therefore, it is far-reaching to explore the role of ncRNAs in tumor development and their potential as therapeutic targets. Here, our review outlines the potential of two major ncRNAs, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) as diagnostic and prognostic biomarkers as well as targets for new therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Ruonan Li
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuxin Ji
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Ruyin Ye
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Department of Life Sciences, Bengbu Medical University, Bengbu, Anhui, China
| | - Guohui Tang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, China
- Department of Life Sciences, Bengbu Medical University, Bengbu, Anhui, China
| | - Wenrui Wang
- Department of Life Sciences, Bengbu Medical University, Bengbu, Anhui, China
| | - Changjie Chen
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
| | - Qingling Yang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
49
|
Lyu K, Tang B, Huang B, Xu Z, Liu T, Fang R, Li Y, Chen Y, Chen L, Zhang M, Chen L, Lei W. Exosomal circPVT1 promotes angiogenesis in laryngeal cancer by activating the Rap1b-VEGFR2 signaling pathway. Carcinogenesis 2024; 45:642-657. [PMID: 38824399 DOI: 10.1093/carcin/bgae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/29/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024] Open
Abstract
Laryngeal cancer (LC) is the second most common head and neck cancer and has a decreasing 5-year survival rate worldwide. Circular RNAs (circRNAs) regulate cancer development in diverse ways based on their distinct biogenesis mechanisms and expansive regulatory roles. However, currently, there is little research on how exosomal circRNAs are involved in the development of LC. Here, we demonstrated that circPVT1, a circRNA derived from the well-studied long noncoding RNA PVT1, is correlated with disease progression in LC and promotes angiogenesis both in vivo and in vitro. Mechanistically, circPVT1 is loaded into LC cell-secreted exosomes and taken up by vascular epithelium cells. By sponging miR-30c-5p, exosomal circPVT1 promotes Rap1b expression, which dramatically enhances vascular endothelial growth factor receptor 2 and the phosphatidylinositol 3-kinase (PI3K)/AKT pathway activation, ultimately resulting in the induction of angiogenesis. Furthermore, our xenograft models demonstrated that the combination of short hairpin RNA-circPVT1 and cetuximab showed high efficacy in inhibiting tumor growth and angiogenesis. Collectively, these findings uncover a novel mechanism of exosomal circRNA-mediated angiogenesis modulation and provide a preclinical rationale for testing this analogous combination in patients with LC.
Collapse
Affiliation(s)
- Kexing Lyu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Bingjie Tang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
- Department of Otorhinolaryngology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, No. 82, Qinglong Street, Qingyang District, Chengdu, Sichuan 610014, China
| | - Bixue Huang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Zhenglin Xu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Tesi Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Ruihua Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Yun Li
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Yi Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Lin Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Minjuan Zhang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Lifan Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| | - Wenbin Lei
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58# Zhongshan Road II, Guangzhou 510080, China
| |
Collapse
|
50
|
Berezowska S, Keyter M, Bouchaab H, Weissferdt A. Pathology of Surgically Resected Lung Cancers Following Neoadjuvant Therapy. Adv Anat Pathol 2024; 31:324-332. [PMID: 38595110 PMCID: PMC11305625 DOI: 10.1097/pap.0000000000000441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
In around 30% of patients, non-small cell lung cancer is diagnosed at an advanced but resectable stage. Adding systemic therapy has shown clear benefit over surgery alone in locally advanced disease, and currently, chemo-immunotherapy in the adjuvant or neoadjuvant setting is the new standard for patients without targetable mutations. One major advantage of the neoadjuvant approach is the possibility of an immediate evaluation of the treatment effect, highlighting the role of pathology as an important contributor at the forefront of clinical decision-making and research. This review provides a summary and an update on current guidelines for histological evaluation of treatment effect after neoadjuvant therapy, also known as regression grading, and discusses newer data focusing on areas of evolving questions and controversies, such as the gross examination of the tumor and tumor bed, weighted versus unweighted evaluation approaches, discussion of histologic tumor type-specific cut-offs for major pathologic response, assessment of lymph nodes and regression grading after immunotherapy and targeted therapy. As no data or recommendations exist on regression grading of multiple tumor nodules, a practical approach is recommended. Lastly, we will touch on additional tissue biomarkers and summarize recent advances in the ardently discussed field of using circulating tumor DNA for the evaluation of treatment response.
Collapse
Affiliation(s)
- Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology
| | - Mark Keyter
- Department of Laboratory Medicine and Pathology, Institute of Pathology
| | - Hasna Bouchaab
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Annikka Weissferdt
- Department of Pathology and Laboratory Medicine
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|