1
|
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
|
2
|
Mondal SK, Hong CS, Han J, Diergaarde B, Zandberg DP, Whiteside TL. Amlodipine, an L-type Ca2+ channel inhibitor, regulates release of extracellular vesicles from tumor cells. Carcinogenesis 2025; 46:bgaf016. [PMID: 40121518 PMCID: PMC12096003 DOI: 10.1093/carcin/bgaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025] Open
Abstract
Tumor cells produce/release tumor-derived exosomes (TEX) which promote tumor growth, drive immune suppression, and interfere with immune therapies. Amlodipine, a calcium flux inhibitor, may block TEX release by tumor cells. Amlodipine's potential as a drug blocking TEX release was evaluated. We measured tumor growth, TEX numbers, phenotype, and molecular content in murine SCCVII and human cancer cell lines. Cell lysates and TEX were tested for expression of autophagy-related proteins by western blots (WBs). Tumor growth in mice, histopathology, T-cell infiltrations, and TEX production by SCCVII treated with amlodipine were measured. Numbers and protein content of TEX eluted from tumor explants were studied by flow cytometry and WBs. Amlodipine used in vitro at 0.5-5 µM was nontoxic, did not impair tumor cell viability, reduced cell proliferation, and decreased TEX production. It reduced PD-L1 and Rab11 content of TEX, altered tumor cell size/shape, induced vesicle accumulations in the cytosol, and upregulated expression levels of autophagy-related proteins, ATG7, Beclin-1, and LC3. In vivo, daily treatment of established SCCVII with amlodipine (10 mg/kg) inhibited tumor growth (P < 0.001), increased CD8+ T-cell infiltration into tumor, decreased TEX production, and altered PD-L1, Rab11, and FasL content of TEX. Amlodipine delivered in vitro to tumor cells or in vivo to tumor-bearing mice interferes with tumor growth and TEX production, induces tumor autophagy, reduces circulating TEX numbers, and alters the TEX immunosuppressive signature. Amlodipine emerges as a potentially promising drug for removing immunosuppressive TEX in cancer subjects who are candidates for immune therapies.
Collapse
Affiliation(s)
- Sujan K Mondal
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Suite 1.32, Pittsburgh, PA 15213, United States
| | - Chang-Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Suite 1.32, Pittsburgh, PA 15213, United States
| | - Jie Han
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Suite 1.32, Pittsburgh, PA 15213, United States
| | - Brenda Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh, UPMC Hillman Cancer Center, 5117 Centre Ave, Pittsburgh, PA 15213, United States
| | - Dan P Zandberg
- Department of Medicine, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Pittsburgh, PA 15213, United States
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Suite 1.32, Pittsburgh, PA 15213, United States
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, 5117 Centre Ave, Suite 1.32, Pittsburgh, PA 15213, United States
| |
Collapse
|
3
|
Jiramonai L, Liang XJ, Zhu M. Extracellular Vesicle-Based Strategies for Tumor Immunotherapy. Pharmaceutics 2025; 17:257. [PMID: 40006624 PMCID: PMC11859549 DOI: 10.3390/pharmaceutics17020257] [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: 12/20/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Immunotherapy is one of the most promising approaches for cancer management, as it utilizes the intrinsic immune response to target cancer cells. Normally, the human body uses its immune system as a defense mechanism to detect and eliminate foreign objects, including cancer cells. However, cancers develop a 'switch off' mechanism, known as immune checkpoint proteins, to evade immune surveillance and suppress immune activation. Therefore, significant efforts have been made to develop the strategies for stimulating immune responses against cancers. Among these, the use of extracellular vesicles (EVs) to enhance the anti-tumor immune response has emerged as a particularly promising approach in cancer management. EVs possess several unique properties that elevate the potency in modulating immune responses. This review article provides a comprehensive overview of recent advances in this field, focusing on the strategic usage of EVs to overcome tumor-induced immune tolerance. We discuss the biogenesis and characteristics of EVs, as well as their potential applications in medical contexts. The immune mechanisms within the tumor microenvironment and the strategies employed by cancers to evade immune detection are explored. The roles of EVs in regulating the tumor microenvironment and enhancing immune responses for immunotherapy are also highlighted. Additionally, this article addresses the challenges and future directions for the development of EV-based nanomedicine approaches, aiming to improve cancer immunotherapy outcomes with greater precision and efficacy while minimizing off-target effects.
Collapse
Affiliation(s)
- Luksika Jiramonai
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Kraft A, Kirschner MB, Orlowski V, Ronner M, Bodmer C, Boeva V, Opitz I, Meerang M. Exploring RNA cargo in extracellular vesicles for pleural mesothelioma detection. BMC Cancer 2025; 25:212. [PMID: 39920655 PMCID: PMC11804012 DOI: 10.1186/s12885-025-13617-y] [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: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Pleural Mesothelioma (PM) is a highly aggressive cancer, for which effective early detection remains a challenge due to limited screening options and low sensitivity of biomarkers discovered so far. While extracellular vesicles (EVs) have emerged as promising candidates for blood-based biomarkers, their role in PM has not been studied yet. In this study, we characterized the transcriptomic profile of EVs secreted by PM primary cells and explored their potential as a biomarker source for PM detection. METHODS We collected cell culture supernatant from early-passage PM cell cultures derived from the pleural effusion of 4 PM patients. EVs were isolated from the supernatant using Qiagen exoEasy Maxi kit. RNA isolation from EVs was done using the mirVana PARIS kit. Finally, single-end RNA sequencing was done with Illumina Novaseq 6000. RESULTS We identified a range of RNA species expressed in EVs secreted by PM cells, including protein-coding RNA (80%), long non-coding RNA (13%), pseudogenes (4.5%), and short non-coding RNA (1.6%). We detected a subset of genes associated with the previously identified epithelioid (32 genes) and sarcomatoid molecular components (36 genes) in PM-EVs. To investigate whether these markers could serve as biomarkers for PM detection in blood, we compared the RNA content of PM-EVs with the cargo of EVs isolated from the plasma of healthy donors (publicly available data). Majority of upregulated genes in PM-EVs were protein-coding and long non-coding RNAs. Interestingly, 25 of them were the sarcomatoid and epithelioid marker genes. Finally, functional analysis revealed that the PM-EV RNA cargo was associated with Epithelial-Mesenchymal transition, glycolysis, and hypoxia. CONCLUSIONS This is the first study to characterize the transcriptomic profile of EVs secreted by PM primary cell cultures, demonstrating their potential as biomarker source for early detection. Further investigation of the functional role of PM-EVs will provide new insights into disease biology and therapeutic avenues.
Collapse
Affiliation(s)
- Agnieszka Kraft
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Michaela B Kirschner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Vanessa Orlowski
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Manuel Ronner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Caroline Bodmer
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- UMR 8104, UMR-S1016, Cochin InstituteCNRSParis Descartes University, Inserm U1016, 75014, Paris, France
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mayura Meerang
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Whiteside TL. Biology of extracellular vesicles and the potential of tumor-derived vesicles for subverting immunotherapy of cancer. J Immunother Cancer 2025; 13:e010376. [PMID: 39855711 PMCID: PMC11759217 DOI: 10.1136/jitc-2024-010376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are produced by all living cells and are present in all body fluids. EVs are heterogeneous in size, biogenesis, molecular/genetic content and functions. They constitute a part of the intercellular communication system. Among them, a subset of small EVs (sEVs) (30-150 nm) originating in the tumor cell endosomes and often referred to as "tumor cell-derived exosomes" have been of special interest. Tumors have adapted sEV they produce to promoting their own survival. Plasma of patients with cancer contains variably elevated numbers of tumor-derived sEV called "TEX," which differ from circulating sEV produced by non-malignant cells by the immunosuppressive phenotype and the molecular/genetic content. Immunosuppressive molecular profiles and abilities to signal, enter and functionally reprogram a variety of recipient cells enable TEX to exert pro-tumor effects that promote tumor resistance to immunotherapy. This review describes phenotypic and functional attributes of TEX that underline their reprogramming capabilities. It also considers mechanisms responsible for TEX pro-tumor activities and the potential significance of TEX signaling for responses of patients with cancer to immune therapies.
Collapse
|
6
|
Naranjo NM, Kennedy A, Testa A, Verrillo CE, Altieri AD, Kean R, Hooper DC, Yu J, Zhao J, Abinader O, Pickles MW, Hawkins A, Kelly WK, Mitra R, Languino LR. Neuroendocrine gene subsets are uniquely dysregulated in prostate adenocarcinoma. Cancer Biol Ther 2024; 25:2364433. [PMID: 38926911 PMCID: PMC11212568 DOI: 10.1080/15384047.2024.2364433] [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: 03/22/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Prostate cancer has heterogeneous growth patterns, and its prognosis is the poorest when it progresses to a neuroendocrine phenotype. Using bioinformatic analysis, we evaluated RNA expression of neuroendocrine genes in a panel of five different cancer types: prostate adenocarcinoma, breast cancer, kidney chromophobe, kidney renal clear cell carcinoma and kidney renal papillary cell carcinoma. Our results show that specific neuroendocrine genes are significantly dysregulated in these tumors, suggesting that they play an active role in cancer progression. Among others, synaptophysin (SYP), a conventional neuroendocrine marker, is upregulated in prostate adenocarcinoma (PRAD) and breast cancer (BRCA). Our analysis shows that SYP is enriched in small extracellular vesicles (sEVs) derived from plasma of PRAD patients, but it is absent in sEVs derived from plasma of healthy donors. Similarly, classical sEV markers are enriched in sEVs derived from plasma of prostate cancer patients, but weakly detectable in sEVs derived from plasma of healthy donors. Overall, our results pave the way to explore new strategies to diagnose these diseases based on the neuroendocrine gene expression in patient tumors or plasma sEVs.
Collapse
Affiliation(s)
- Nicole M. Naranjo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anne Kennedy
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anna Testa
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cecilia E. Verrillo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adrian D. Altieri
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - D. Craig Hooper
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jindan Yu
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan Zhao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Oliver Abinader
- Division of Biostatistics and Bioinformatics, Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maxwell W. Pickles
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Hawkins
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - William K. Kelly
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ramkrishna Mitra
- Division of Biostatistics and Bioinformatics, Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia R. Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Mir R, Baba SK, Elfaki I, Algehainy N, Alanazi MA, Altemani FH, Tayeb FJ, Barnawi J, Husain E, Bedaiwi RI, Albalawi IA, Alhujaily M, Mir MM, Almotairi R, Alatwi HE, Albalawi AD. Unlocking the Secrets of Extracellular Vesicles: Orchestrating Tumor Microenvironment Dynamics in Metastasis, Drug Resistance, and Immune Evasion. J Cancer 2024; 15:6383-6415. [PMID: 39513123 PMCID: PMC11540496 DOI: 10.7150/jca.98426] [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: 05/14/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Extracellular vehicles (EVs) are gaining increasing recognition as central contributors to the intricate landscape of the tumor microenvironment (TME). This manuscript provides an extensive examination of the multifaceted roles played by EVs in shaping the TME, with a particular emphasis on their involvement in metastasis, drug resistance, and immune evasion. Metastasis, the process by which cancer cells disseminate to distant sites, remains a formidable challenge in cancer management. EVs, encompassing exosomes and microvesicles, have emerged as critical participants in this cascade of events. They facilitate the epithelial-to-mesenchymal transition (EMT), foster pre-metastatic niche establishment, and enhance the invasive potential of cancer cells. This manuscript delves into the intricate molecular mechanisms underpinning these processes, underscoring the therapeutic potential of targeting EVs to impede metastasis. Drug resistance represents a persistent impediment to successful cancer treatment. EVs are instrumental in intrinsic and acquired drug resistance, acting as mediators of intercellular communication. They ferry molecules like miRNAs and proteins, which confer resistance to conventional chemotherapy and targeted therapies. This manuscript scrutinizes the diverse strategies employed by EVs in propagating drug resistance while also considering innovative approaches involving EV-based drug delivery systems to counteract this phenomenon. Immune evasion is a hallmark of cancer, and EVs are central in sculpting the immunosuppressive milieu of the TME. Tumor-derived EVs thwart immune responses through various mechanisms, including T cell dysfunction induction, the expansion of regulatory T cells (Tregs), and polarization of macrophages towards an immunosuppressive phenotype. In addition, the manuscript explores the diagnostic potential of EVs as biomarkers and their role as therapeutic agents in immune checkpoint blockade therapies. This manuscript provides a comprehensive overview of EV's pivotal role in mediating intricate interactions within the TME, ultimately influencing cancer progression and therapeutic outcomes. A profound understanding of EV-mediated processes in metastasis, drug resistance, and immune evasion opens up promising avenues for developing innovative therapeutic strategies and identifying valuable biomarkers in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Sadaf Khursheed Baba
- Watson Crick Center for Molecular Medicine, Islamic University of Science and Technology, J & K, India
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Faris Jamal Tayeb
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Jameel Barnawi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Eram Husain
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Ruqaiah I Bedaiwi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Muhanad Alhujaily
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Mohammad Muzaffar Mir
- Department of Biochemistry, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Reema Almotairi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Hanan E. Alatwi
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | | |
Collapse
|
8
|
Whiteside TL. Tumor-derived Exosomes and Antitumor Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:923-931. [PMID: 39284119 PMCID: PMC11951267 DOI: 10.4049/jimmunol.2400335] [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: 06/10/2024] [Accepted: 08/01/2024] [Indexed: 11/13/2024]
Abstract
Cancer immunotherapy, including immune checkpoint blockade, has been approved for treatment of patients with many cancer types. However, some patients fail to respond to immunotherapy, and emerging evidence indicates that tumor-derived exosomes (TEX) play a major role in reprogramming the host immune cells by inducing their dysfunction. Focusing on effector T cells, this review illustrates mechanisms of suppression that TEX use, thus promoting tumor escape from the host immune system. TEX carry multiple suppressive signals that drive T cell dysfunction and convert the tumor microenvironment into "an immune desert" in which activated T cells either die or are reprogrammed to mediate protumor functions. The reprogrammed T cells produce a new crop of CD3+ immunoinhibitory exosomes that further amplify suppression mediated by TEX. The result is a profound depletion of antitumor immune effector cells that reflects the defective immune competence of the cancer patient and partly explains why TEX are a significant barrier for cancer immunotherapy.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA; and UPMC Hillman Cancer Center, Pittsburgh, PA
| |
Collapse
|
9
|
Zandberg DP, Hong CS, Swartz A, Hsieh R, Anderson J, Ferris RL, Diergaarde B, Whiteside TL. Small extracellular vesicles as biomarkers of response in recurrent/metastatic HNSCC patients treated with immunotherapy. BJC REPORTS 2024; 2:70. [PMID: 39281316 PMCID: PMC11390474 DOI: 10.1038/s44276-024-00096-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/12/2024] [Accepted: 08/28/2024] [Indexed: 09/18/2024]
Abstract
Background Biomarkers that effectively predict response to anti-PD-1 mAb therapy in cancer patients are an unmet need. We evaluated the utility of small extracellular vesicles (sEV) as biomarkers of response to immunotherapy in recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) patients. Methods Plasma sEV were isolated from 24 R/M HNSCC patients prior to immunotherapy initiation. sEV were separated by immune capture into T cell-derived CD3(+) and tumor-enriched CD3(-) subsets. Stimulatory and suppressive profiles of CD3(-) sEV were determined by on-bead flow cytometry. Differences were assessed using nonparametric tests. Multivariable Cox regression was used to evaluate the relationship with overall (OS) and progression free survival (PFS). Results CD3(-)CD44v3(+) sEV represented the majority of plasma sEV; the T-cell-derived CD3(+) fraction was significantly smaller. High CD3(+) sEV was associated with better OS and PFS. Total CD3(-)CD44v3(+) sEV was not associated with outcome. However, suppressive and stimulatory profiles were associated with OS; the suppressive/stimulatory ratio was associated with best response. Exploration of individual proteins on CD3(-) sEV showed that high PD-L1 and high CTLA-4 were associated with better outcomes. Conclusions Evaluation of the T cell-derived-CD3(+) and tumor-enriched CD3(-) plasma sEV subsets indicated their potential utility as biomarkers of response to immunotherapy.
Collapse
Affiliation(s)
| | - Chang-Sook Hong
- UPMC Hillman Cancer Center, Pittsburgh, PA USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | | | - Ronan Hsieh
- UPMC Hillman Cancer Center, Pittsburgh, PA USA
| | | | | | - Brenda Diergaarde
- UPMC Hillman Cancer Center, Pittsburgh, PA USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA USA
| | - Theresa L. Whiteside
- UPMC Hillman Cancer Center, Pittsburgh, PA USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
10
|
Skoczylas Ł, Gawin M, Fochtman D, Widłak P, Whiteside TL, Pietrowska M. Immune capture and protein profiling of small extracellular vesicles from human plasma. Proteomics 2024; 24:e2300180. [PMID: 37713108 PMCID: PMC11046486 DOI: 10.1002/pmic.202300180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Extracellular vesicles (EVs), the key players in inter-cellular communication, are produced by all cell types and are present in all body fluids. Analysis of the proteome content is an important approach in structural and functional studies of these vesicles. EVs circulating in human plasma are heterogeneous in size, cellular origin, and functions. This heterogeneity and the potential presence of contamination with plasma components such as lipoprotein particles and soluble plasma proteins represent a challenge in profiling the proteome of EV subsets by mass spectrometry. An immunocapture strategy prior to mass spectrometry may be used to isolate a homogeneous subpopulation of small EVs (sEV) with a specific endocytic origin from plasma or other biofluids. Immunocapture selectively separates EV subpopulations in biofluids based on the presence of a unique protein carried on the vesicle surface. The advantages and disadvantages of EV immune capture as a preparative step for mass spectrometry are discussed.
Collapse
Affiliation(s)
- Łukasz Skoczylas
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Marta Gawin
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Daniel Fochtman
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
- Silesian University of Technology, 44-100 Gliwice, Poland
| | - Piotr Widłak
- Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Theresa L. Whiteside
- UPMC Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Monika Pietrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| |
Collapse
|
11
|
Shi Y, Yao F, Yin Y, Wu C, Xia D, Zhang K, Jin Z, Liu X, He J, Zhang Z. Extracellular vesicles derived from immune cells: Role in tumor therapy. Int Immunopharmacol 2024; 133:112150. [PMID: 38669949 DOI: 10.1016/j.intimp.2024.112150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Extracellular vesicles (EVs), which have a lipid nano-sized structure, are known to contain the active components of parental cells and play a crucial role in intercellular communication. The progression and metastasis of tumors are influenced by EVs derived from immune cells, which can simultaneously stimulate and suppress immune responses. In the past few decades, there has been a considerable focus on EVs due to their potential in various areas such as the development of vaccines, delivering drugs, making engineered modifications, and serving as biomarkers for diagnosis and prognosis. This review focuses on the substance information present in EVs derived from innate and adaptive immune cells, their effects on the immune system, and their applications in cancer treatment. While there are still challenges to overcome, it is important to explore the composition of immune cells released vesicles and their potential therapeutic role in tumor therapy. The review also highlights the current limitations and future prospects in utilizing EVs for treatment purposes.
Collapse
Affiliation(s)
- Yuanyuan Shi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Fei Yao
- Department of Oncology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Yao Yin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Chen Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Desong Xia
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Keyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Ze Jin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China.
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China.
| | - Zhikun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China; The Second Affiliated Hospital of Guangxi Medical University, Nanning 530023, China.
| |
Collapse
|
12
|
Andre M, Caobi A, Miles JS, Vashist A, Ruiz MA, Raymond AD. Diagnostic potential of exosomal extracellular vesicles in oncology. BMC Cancer 2024; 24:322. [PMID: 38454346 PMCID: PMC10921614 DOI: 10.1186/s12885-024-11819-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024] Open
Abstract
Liquid biopsy can detect circulating cancer cells or tumor cell-derived DNA at various stages of cancer. The fluid from these biopsies contains extracellular vesicles (EVs), such as apoptotic bodies, microvesicles, exomeres, and exosomes. Exosomes contain proteins and nucleic acids (DNA/RNA) that can modify the microenvironment and promote cancer progression, playing significant roles in cancer pathology. Clinically, the proteins and nucleic acids within the exosomes from liquid biopsies can be biomarkers for the detection and prognosis of cancer. We review EVs protein and miRNA biomarkers identified for select cancers, specifically melanoma, glioma, breast, pancreatic, hepatic, cervical, prostate colon, and some hematological malignancies. Overall, this review demonstrates that EV biomolecules have great potential to expand the diagnostic and prognostic biomarkers used in Oncology; ultimately, EVs could lead to earlier detection and novel therapeutic targets. Clinical implicationsEVs represent a new paradigm in cancer diagnostics and therapeutics. The potential use of exosomal contents as biomarkers for diagnostic and prognostic indicators may facilitate cancer management. Non-invasive liquid biopsy is helpful, especially when the tumor is difficult to reach, such as in pancreatic adenocarcinoma. Moreover, another advantage of using minimally invasive liquid biopsy is that monitoring becomes more manageable. Identifying tumor-derived exosomal proteins and microRNAs would allow a more personalized approach to detecting cancer and improving treatment.
Collapse
Affiliation(s)
- Mickensone Andre
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA
| | - Allen Caobi
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA
| | - Jana S Miles
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA
| | - Arti Vashist
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA
| | - Marco A Ruiz
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA
- Medical Oncology, Baptist Health Miami Cancer Institute, Miami, 33176, FL, USA
| | - Andrea D Raymond
- Herbert Wertheim College of Medicine at, Department of Immunology and Nanomedicine, Florida International University, Miami, 33199, FL, USA.
| |
Collapse
|
13
|
Lattmann E, Räss L, Tognetti M, Gómez JMM, Lapaire V, Bruderer R, Reiter L, Feng Y, Steinmetz LM, Levesque MP. Size-exclusion chromatography combined with DIA-MS enables deep proteome profiling of extracellular vesicles from melanoma plasma and serum. Cell Mol Life Sci 2024; 81:90. [PMID: 38353833 PMCID: PMC10867102 DOI: 10.1007/s00018-024-05137-y] [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: 04/23/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
Extracellular vesicles (EVs) are important players in melanoma progression, but their use as clinical biomarkers has been limited by the difficulty of profiling blood-derived EV proteins with high depth of coverage, the requirement for large input amounts, and complex protocols. Here, we provide a streamlined and reproducible experimental workflow to identify plasma- and serum- derived EV proteins of healthy donors and melanoma patients using minimal amounts of sample input. SEC-DIA-MS couples size-exclusion chromatography to EV concentration and deep-proteomic profiling using data-independent acquisition. From as little as 200 µL of plasma per patient in a cohort of three healthy donors and six melanoma patients, we identified and quantified 2896 EV-associated proteins, achieving a 3.5-fold increase in depth compared to previously published melanoma studies. To compare the EV-proteome to unenriched blood, we employed an automated workflow to deplete the 14 most abundant proteins from plasma and serum and thereby approximately doubled protein group identifications versus native blood. The EV proteome diverged from corresponding unenriched plasma and serum, and unlike the latter, separated healthy donor and melanoma patient samples. Furthermore, known melanoma markers, such as MCAM, TNC, and TGFBI, were upregulated in melanoma EVs but not in depleted melanoma plasma, highlighting the specific information contained in EVs. Overall, EVs were significantly enriched in intact membrane proteins and proteins related to SNARE protein interactions and T-cell biology. Taken together, we demonstrated the increased sensitivity of an EV-based proteomic workflow that can be easily applied to larger melanoma cohorts and other indications.
Collapse
Affiliation(s)
- Evelyn Lattmann
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Luca Räss
- Biognosys AG, Schlieren, Switzerland
| | | | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | - Valérie Lapaire
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| | | | | | | | - Lars M Steinmetz
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
| |
Collapse
|
14
|
Liao Y, Zhang Z, Ouyang L, Mi B, Liu G. Engineered Extracellular Vesicles in Wound Healing: Design, Paradigms, and Clinical Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307058. [PMID: 37806763 DOI: 10.1002/smll.202307058] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/20/2023] [Indexed: 10/10/2023]
Abstract
The severe quality of life and economic burden imposed by non-healing skin wounds, infection risks, and treatment costs are affecting millions of patients worldwide. To mitigate these challenges, scientists are relentlessly seeking effective treatment measures. In recent years, extracellular vesicles (EVs) have emerged as a promising cell-free therapy strategy, attracting extensive attention from researchers. EVs mediate intercellular communication, possessing excellent biocompatibility and stability. These features make EVs a potential tool for treating a plethora of diseases, including those related to wound repair. However, there is a growing focus on the engineering of EVs to overcome inherent limitations such as low production, relatively fixed content, and targeting capabilities of natural EVs. This engineering could improve both the effectiveness and specificity of EVs in wound repair treatments. In light of this, the present review will introduce the latest progress in the design methods and experimental paradigms of engineered EVs applied in wound repair. Furthermore, it will comprehensively analyze the current clinical research status and prospects of engineered EVs within this field.
Collapse
Affiliation(s)
- Yuheng Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zhenhe Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Lizhi Ouyang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| |
Collapse
|
15
|
Łuczak MW, Dżaman K, Zaręba Ł, Czerwaty K, Siewiera J, Głuszko A, Olszewska E, Brzost J, Kantor I, Szczepański MJ, Ludwig N. HMGB1 Carried by Small Extracellular Vesicles Potentially Plays a Role in Promoting Acquired Middle Ear Cholesteatoma. Diagnostics (Basel) 2023; 13:3469. [PMID: 37998605 PMCID: PMC10669961 DOI: 10.3390/diagnostics13223469] [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: 09/20/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Cholesteatoma is a specific medical condition involving the abnormal, non-cancerous growth of skin-like tissue in the middle ear, potentially leading to a collection of debris and even infections. The receptor for advanced glycation (RAGE) and its ligand, high-mobility box 1 (HMGB1), are both known to be overexpressed in cholesteatoma and play a potential role in the pathogenesis of the disease. In this study, we investigated the role of small extracellular vesicles (sEVs) in carrying HMGB1 and inducing disease-promoting effects in cholesteatoma. No significant differences in the concentration of isolated sEVs in the plasma of cholesteatoma patients (n = 17) and controls (n = 22) were found (p > 0.05); however, cholesteatoma-derived sEVs carried significantly higher levels of HMGB1 (p < 0.05). In comparison to sEVs isolated from the plasma of controls, cholesteatoma-derived sEVs significantly enhanced keratinocyte proliferation and IL-6 production (p < 0.05), potentially by engaging multiple activation pathways including MAPKp44/p42, STAT3, and the NF-κB pathway. Thus, HMGB1(+) sEVs emerge as a novel factor potentially promoting cholesteatoma progression.
Collapse
Affiliation(s)
- Michał W. Łuczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02906, USA;
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (Ł.Z.); (A.G.)
| | - Karolina Dżaman
- Department of Otolaryngology, Centre of Postgraduate Medical Education, 02-097 Warsaw, Poland; (K.D.); (K.C.); (I.K.)
| | - Łukasz Zaręba
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (Ł.Z.); (A.G.)
| | - Katarzyna Czerwaty
- Department of Otolaryngology, Centre of Postgraduate Medical Education, 02-097 Warsaw, Poland; (K.D.); (K.C.); (I.K.)
| | - Jacek Siewiera
- Department of Hyperbaric Medicine, Military Institute of Medicine-National Research Institute, 00-902 Warsaw, Poland;
| | - Alicja Głuszko
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (Ł.Z.); (A.G.)
| | - Ewa Olszewska
- Department of Otolaryngology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Jacek Brzost
- Department of Otolaryngology, The Children’s Memorial Health Institute, 00-328 Warsaw, Poland;
| | - Ireneusz Kantor
- Department of Otolaryngology, Centre of Postgraduate Medical Education, 02-097 Warsaw, Poland; (K.D.); (K.C.); (I.K.)
| | - Mirosław J. Szczepański
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland; (Ł.Z.); (A.G.)
- Department of Otolaryngology, Centre of Postgraduate Medical Education, 02-097 Warsaw, Poland; (K.D.); (K.C.); (I.K.)
| | - Nils Ludwig
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
16
|
Lee YJ, Chae S, Choi D. Monitoring of single extracellular vesicle heterogeneity in cancer progression and therapy. Front Oncol 2023; 13:1256585. [PMID: 37823055 PMCID: PMC10562638 DOI: 10.3389/fonc.2023.1256585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
Cancer cells actively release lipid bilayer extracellular vesicles (EVs) that affect their microenvironment, favoring their progression and response to extracellular stress. These EVs contain dynamically regulating molecular cargos (proteins and nucleic acids) selected from their parental cells, representing the active biological functionality for cancer progression. These EVs are heterogeneous according to their size and molecular composition and are usually defined based on their biogenetic mechanisms, such as exosomes and ectosomes. Recent single EV detection technologies, such as nano-flow cytometry, have revealed the dynamically regulated molecular diversity within bulk EVs, indicating complex EV heterogeneity beyond classical biogenetic-based EV subtypes. EVs can be changed by internal oncogenic transformation or external stress such as chemotherapy. Among the altered combinations of EV subtypes, only a specific set of EVs represents functional molecular cargo, enabling cancer progression and immune modulation in the tumor microenvironment through their altered targeting efficiency and specificity. This review covers the heterogeneity of EVs discovered by emerging single EV analysis technologies, which reveal the complex distribution of EVs affected by oncogenic transformation and chemotherapy. Encouragingly, these unique molecular signatures in individual EVs indicate the status of their parental cancer cells. Thus, precise molecular profiling of circulating single EVs would open new areas for in-depth monitoring of the cancer microenvironment and shed new light on non-invasive diagnostic approaches using liquid biopsy.
Collapse
Affiliation(s)
| | | | - Dongsic Choi
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan, Chungcheongnam, Republic of Korea
| |
Collapse
|
17
|
Matas-Nadal C, Bech-Serra JJ, Gatius S, Gomez X, Ribes-Santolaria M, Guasch-Vallés M, Pedraza N, Casanova JM, Gómez CDLT, Garí E, Aguayo-Ortiz RS. Biomarkers found in the tumor interstitial fluid may help explain the differential behavior among keratinocyte carcinomas. Mol Cell Proteomics 2023; 22:100547. [PMID: 37059366 DOI: 10.1016/j.mcpro.2023.100547] [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: 07/26/2022] [Revised: 04/01/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023] Open
Abstract
Basal Cell Carcinomas (BCC) and cutaneous Squamous Cell Carcinomas (SCC) are the most frequent types of cancer, and both originate from the keratinocyte transformation, giving rise to the group of tumors called keratinocyte carcinomas (KC). The invasive behavior is different in each group of KC and may be influenced by their tumor microenvironment. The principal aim of the study is to characterize the protein profile of the Tumor Interstitial Fluid (TIF) of KC to evaluate changes in the microenvironment that could be associated with their different invasive and metastatic capabilities. We obtained TIF from 27 skin biopsies and conducted a label-free quantitative proteomic analysis comparing 7 BCCs, 16 SCCs, and 4 Normal Skins. A total of 2945 proteins were identified, 511 of them quantified in more than half of the samples of each tumoral type. The proteomic analysis revealed differentially expressed TIF-proteins that could explain the different metastatic behavior in both KC. In detail, the SCC samples disclosed an enrichment of proteins related to cytoskeleton, such as Stratafin and Ladinin1. Previous studies found their up-regulation positively correlated with tumor progression. Furthermore, the TIF of SCC samples was enriched with the cytokines S100A8/S100A9. These cytokines influence the metastatic output in other tumors through the activation of NF-kB signaling. According to this, we observed a significant increase in nuclear NF-kB subunit p65 in SCCs but not in BCCs. In addition, the TIF of both tumors was enriched with proteins involved in the immune response, highlighting the relevance of this process in the composition of the tumor environment. Thus, the comparison of the TIF composition of both KC provides the discovery of a new set of differential biomarkers. Among them, secreted cytokines such as S100A9 may help explain the higher aggressiveness of SCCs, while Cornulin is a specific biomarker for BCCs. Finally, the proteomic landscape of TIF provides key information on tumor growth and metastasis, which can contribute to the identification of clinically applicable biomarkers that may be used in the diagnosis of KC, as well as therapeutic targets.
Collapse
Affiliation(s)
- Clara Matas-Nadal
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dermatology department. Hospital Santa Caterina, Salt, Girona.
| | - Joan J Bech-Serra
- Proteomics Unit, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Sònia Gatius
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Servei d'anatomia patològica, Hospital Universitari Arnau de Vilanova, Lleida
| | - Xavier Gomez
- Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida
| | - Marina Ribes-Santolaria
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida
| | - Marta Guasch-Vallés
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida
| | - Neus Pedraza
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida
| | - Josep M Casanova
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida; Servei de Dermatologia, Hospital Universitari Arnau de Vilanova, Lleida
| | | | - Eloi Garí
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida
| | - Rafael S Aguayo-Ortiz
- Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida); Dept. Ciències Mèdiques Bàsiques. Facultat de Medicina. Universitat de Lleida; Servei de Dermatologia, Hospital Universitari Arnau de Vilanova, Lleida; Cell cycle lab. Institut de Recerca Biomèdica de Lleida (IRB Lleida)
| |
Collapse
|
18
|
Hua X, Zhu Q, Liu Y, Zhou S, Huang P, Li Q, Liu S. A double tangential flow filtration-based microfluidic device for highly efficient separation and enrichment of exosomes. Anal Chim Acta 2023; 1258:341160. [PMID: 37087290 DOI: 10.1016/j.aca.2023.341160] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Recently, exosomes have been recognized as important disease biomarkers due to the essential roles they played in disease development. Nevertheless, the highly efficient isolation and enrichment of exosomes from complex body fluids continues to hinder the research and application of exosomes for clinical use. In this work, we developed a double tangential flow filtration-based microfluidic device for exosome isolation from cell supernatants and human serum. The microfluidic device contained two modules. Each module included two polymethylmethacrylate (PMMA) plates with symmetrical serpentine channels and a nanoporous membrane with 200 nm or 30 nm pore diameter and was used to separate larger vesicles, exosomes and free biomolecules. The design of double tangential flow filtration in symmetrical serpentine channels largely increased the contact area between the filtrate and the nanoporous membranes, thus improved the separation efficiency and prevented the clogging of the membrane. Compared with standard separation method, i.e. ultracentrifugation (UC), the microfluidic chip-based separation (Chip) of exosomes showed the advantages of much lower instrumental cost, lower consumable cost, shorter time (<120 min), higher purity (82.8%) and significantly higher recovery rate (77.8%). In addition, due to the label-free separation, the microfluidic device-collected exosomes could be directly used for downstream analysis such as proteomics analysis. The proteomics analysis results of exosomes isolated from the sera of clinical patients with different diseases by the chip revealed richer disease-related information comparing with those exosomes isolated by UC, demonstrating the good practicability of this chip for future clinical research and applications.
Collapse
|
19
|
Surman M, Jankowska U, Wilczak M, Przybyło M. Similarities and Differences in the Protein Composition of Cutaneous Melanoma Cells and Their Exosomes Identified by Mass Spectrometry. Cancers (Basel) 2023; 15:cancers15041097. [PMID: 36831440 PMCID: PMC9954195 DOI: 10.3390/cancers15041097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Intercellular transport of proteins mediated by extracellular vesicles (EVs)-exosomes and ectosomes-is one of the factors facilitating carcinogenesis. Therefore, the research on protein cargo of melanoma-derived EVs may provide a better understanding of the mechanisms involved in melanoma progression and contribute to the development of alternative biomarkers. Proteomic data on melanoma-derived EVs are very limited. The shotgun nanoLC-MS/MS approach was applied to analyze the protein composition of primary (WM115, WM793) and metastatic (WM266-4, WM1205Lu) cutaneous melanoma cells and exosomes released by them. All cells secreted homogeneous populations of exosomes that shared a characteristic set of proteins. In total, 3514 and 1234 unique proteins were identified in melanoma cells and exosomes, respectively. Gene ontology analysis showed enrichment in several cancer-related categories, including cell proliferation, migration, negative regulation of apoptosis, and angiogenesis. The obtained results broaden our knowledge on the role of selected proteins in exosome biology, as well as their functional role in the development and progression of cutaneous melanoma. The results may also inspire future studies on the clinical potential of exosomes.
Collapse
Affiliation(s)
- Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland
- Correspondence: ; Tel.: +48-12-664-6462
| |
Collapse
|
20
|
Whiteside TL. Evaluating tumor cell- and T cell-derived extracellular vesicles as potential biomarkers of cancer and immune cell competence. Expert Rev Mol Diagn 2023; 23:109-122. [PMID: 36787282 PMCID: PMC9998373 DOI: 10.1080/14737159.2023.2178902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) produced by tumors, also called tumor-derived exosomes (TEX), have been implicated in inducing immune cell suppression in vitro and in vivo. The development of a novel category of noninvasive biomarkers for precision oncology remains an unmet need, and TEX emerge as a promising liquid tumor biopsy component. AREAS COVERED TEX play a critical role in monitoring cancer presence/progression and in reprograming of anti-tumor effector T cells to producers of EVs with pro-tumor activity. TEX are a subset of circulating EVs. Their separation by immune capture from EVs derived from nonmalignant cells allows for TEX phenotypic/functional assessments. TEX cross-talking with CD3(+) T cells induce the release of CD3(+) small EV (sEV), whose cargo of suppressor proteins resembles that of TEX and further contributes to cancer-induced immune suppression. While TEX recapitulate the genetic/molecular phenotype of tumor cells, CD3(+) sEV might serve as 'T cell liquid biopsy.' EXPERT OPINION Preclinical explorations of the role in cancer body fluids of TEX and CD3(+) sEV as cancer biomarkers suggest that these EV subsets may qualify as liquid tumor biopsy noninvasive components in the near future. Their potential to simultaneously serve as noninvasive liquid tumor biopsy and T cell biopsy remains to be validated in future clinical trials.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Departments of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Medhin LB, Beasley AB, Warburton L, Amanuel B, Gray ES. Extracellular vesicles as a liquid biopsy for melanoma: Are we there yet? Semin Cancer Biol 2023; 89:92-98. [PMID: 36706847 DOI: 10.1016/j.semcancer.2023.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Melanoma is the most aggressive form of skin cancer owing to its high propensity to metastasise in distant organs and develop resistance to treatment. The scarce treatment options available for melanoma underscore the need for biomarkers to guide treatment decisions. In this context, an attractive alternative to overcome the limitations of repeated tissue sampling is the analysis of peripheral blood samples, referred to as 'liquid biopsy'. In particular, the analysis of extracellular vesicles (EVs) has emerged as a promising candidate due to their role in orchestrating cancer dissemination, immune modulation, and drug resistance. As we gain insights into the role of EVs in cancer and melanoma their potential for clinical use is becoming apparent. Herein, we critically summarise the current evidence supporting EVs as biomarkers for melanoma diagnosis, prognostication, therapy response prediction, and drug resistance. EVs are proposed as a candidate biomarker for predicting therapeutic response to immune checkpoint inhibition. However, to realise the potential of EV analysis for clinical decision-making strong clinical validation is required, underscoring the need for further research in this area.
Collapse
Affiliation(s)
- Lidia B Medhin
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia
| | - Aaron B Beasley
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia
| | - Lydia Warburton
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia; Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Australia
| | - Benhur Amanuel
- School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia; Department of Anatomical Pathology PathWest, QEII Medical Centre, Nedlands WA 6009, Australia
| | - Elin S Gray
- Centre for Precision Health, Edith Cowan University, Joondalup WA 6027, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia.
| |
Collapse
|
22
|
Whiteside T, Zarour HM. In memoriam: Soldano Ferrone, MD, PhD (1940-2023). J Immunother Cancer 2023; 11:jitc-2023-006761. [PMID: 36717185 PMCID: PMC9887713 DOI: 10.1136/jitc-2023-006761] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Affiliation(s)
- Theresa Whiteside
- Pathology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Hassane M Zarour
- Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
23
|
Benito-Martín A, Jasiulionis MG, García-Silva S. Extracellular vesicles and melanoma: New perspectives on tumor microenvironment and metastasis. Front Cell Dev Biol 2023; 10:1061982. [PMID: 36704194 PMCID: PMC9871288 DOI: 10.3389/fcell.2022.1061982] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Secreted extracellular vesicles (EVs) are lipid bilayer particles without functional nucleus naturally released from cells which constitute an intercellular communication system. There is a broad spectrum of vesicles shed by cells based on their physical properties such as size (small EVs and large EVs), biogenesis, cargo and functions, which provide an increasingly heterogenous landscape. In addition, they are involved in multiple physiological and pathological processes. In cancer, EV release is opted by tumor cells as a beneficial process for tumor progression. Cutaneous melanoma is a cancer that originates from the melanocyte lineage and shows a favorable prognosis at early stages. However, when melanoma cells acquire invasive capacity, it constitutes the most aggressive and deadly skin cancer. In this context, extracellular vesicles have been shown their relevance in facilitating melanoma progression through the modulation of the microenvironment and metastatic spreading. In agreement with the melanosome secretory capacity of melanocytes, melanoma cells display an enhanced EV shedding activity that has contributed to the utility of melanoma models for unravelling EV cargo and functions within a cancer scenario. In this review, we provide an in-depth overview of the characteristics of melanoma-derived EVs and their role in melanoma progression highlighting key advances and remaining open questions in the field.
Collapse
Affiliation(s)
- Alberto Benito-Martín
- Facultad de Medicina, Unidad de Investigación Biomédica, Universidad Alfonso X El Sabio (UAX), Villanueva de la Cañada, Spain,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| | - Miriam Galvonas Jasiulionis
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| | - Susana García-Silva
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| |
Collapse
|
24
|
Zhang M, Lu Y, Wang L, Mao Y, Hu X, Chen Z. Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors. Cancers (Basel) 2022; 15:cancers15010100. [PMID: 36612097 PMCID: PMC9817817 DOI: 10.3390/cancers15010100] [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: 11/05/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are important mediators of communication between tumor cells and normal cells. These vesicles are rich in a variety of contents such as RNA, DNA, and proteins, and can be involved in angiogenesis, epithelial-mesenchymal transition, the formation of pre-metastatic ecological niches, and the regulation of the tumor microenvironment. Small extracellular vesicles (sEVs) are a type of EVs. Currently, the main treatments for urological tumors are surgery, radiotherapy, and targeted therapy. However, urological tumors are difficult to diagnose and treat due to their high metastatic rate, tendency to develop drug resistance, and the low sensitivity of liquid biopsies. Numerous studies have shown that sEVs offer novel therapeutic options for tumor treatment, such as tumor vaccines and tumor drug carriers. sEVs have attracted a great deal of attention owing to their contribution to in intercellular communication, and as novel biomarkers, and role in the treatment of urological tumors. This article reviews the research and applications of sEVs in the diagnosis and treatment of urological tumors.
Collapse
Affiliation(s)
- Mengting Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Yukang Lu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Yiping Mao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xinyi Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhiping Chen
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Correspondence: ; Tel.: +86-150-8373-7280
| |
Collapse
|
25
|
Temperini ME, Di Giacinto F, Romanò S, Di Santo R, Augello A, Polito R, Baldassarre L, Giliberti V, Papi M, Basile U, Niccolini B, Krasnowska EK, Serafino A, De Spirito M, Di Gaspare A, Ortolani M, Ciasca G. Antenna-enhanced mid-infrared detection of extracellular vesicles derived from human cancer cell cultures. J Nanobiotechnology 2022; 20:530. [PMID: 36514065 PMCID: PMC9746222 DOI: 10.1186/s12951-022-01693-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Extracellular Vesicles (EVs) are sub-micrometer lipid-bound particles released by most cell types. They are considered a promising source of cancer biomarkers for liquid biopsy and personalized medicine due to their specific molecular cargo, which provides biochemical information on the state of parent cells. Despite this potential, EVs translation process in the diagnostic practice is still at its birth, and the development of novel medical devices for their detection and characterization is highly required. RESULTS In this study, we demonstrate mid-infrared plasmonic nanoantenna arrays designed to detect, in the liquid and dry phase, the specific vibrational absorption signal of EVs simultaneously with the unspecific refractive index sensing signal. For this purpose, EVs are immobilized on the gold nanoantenna surface by immunocapture, allowing us to select specific EV sub-populations and get rid of contaminants. A wet sample-handling technique relying on hydrophobicity contrast enables effortless reflectance measurements with a Fourier-transform infrared (FTIR) spectro-microscope in the wavelength range between 10 and 3 µm. In a proof-of-principle experiment carried out on EVs released from human colorectal adenocarcinoma (CRC) cells, the protein absorption bands (amide-I and amide-II between 5.9 and 6.4 µm) increase sharply within minutes when the EV solution is introduced in the fluidic chamber, indicating sensitivity to the EV proteins. A refractive index sensing curve is simultaneously provided by our sensor in the form of the redshift of a sharp spectral edge at wavelengths around 5 µm, where no vibrational absorption of organic molecules takes place: this permits to extract of the dynamics of EV capture by antibodies from the overall molecular layer deposition dynamics, which is typically measured by commercial surface plasmon resonance sensors. Additionally, the described metasurface is exploited to compare the spectral response of EVs derived from cancer cells with increasing invasiveness and metastatic potential, suggesting that the average secondary structure content in EVs can be correlated with cell malignancy. CONCLUSIONS Thanks to the high protein sensitivity and the possibility to work with small sample volumes-two key features for ultrasensitive detection of extracellular vesicles- our lab-on-chip can positively impact the development of novel laboratory medicine methods for the molecular characterization of EVs.
Collapse
Affiliation(s)
- Maria Eleonora Temperini
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185, Rome, Italy
- Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Flavio Di Giacinto
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sabrina Romanò
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Di Santo
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Alberto Augello
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| | - Raffaella Polito
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185, Rome, Italy
| | - Leonetta Baldassarre
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185, Rome, Italy
| | - Valeria Giliberti
- Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Umberto Basile
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168, Rome, Italy
| | - Benedetta Niccolini
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ewa K Krasnowska
- Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Annalucia Serafino
- Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| | - Marco De Spirito
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Di Gaspare
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
- NEST, CNR-Istituto Nanoscienze and Scuola Normale Superiore, Piazza San Silvestro 12, 56127, Pisa, Italy
| | - Michele Ortolani
- Department of Physics, Sapienza University of Rome, Piazzale Aldo Moro 2, 00185, Rome, Italy.
- Center for Life Neuro and Nano Sciences IIT@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Gabriele Ciasca
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy.
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
26
|
Li Y, Zhao W, Fu R, Ma Z, Hu Y, Liu Y, Ding Z. Endoplasmic reticulum stress increases exosome biogenesis and packaging relevant to sperm maturation in response to oxidative stress in obese mice. Reprod Biol Endocrinol 2022; 20:161. [PMID: 36411474 PMCID: PMC9677646 DOI: 10.1186/s12958-022-01031-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Mammalian sperm maturation in the epididymis is mainly modulated by exosomes that are secreted into the epididymal lumen from epididymal epithelial cells (EECs). Exposure to oxidative stress (OS) resulting from being fed a high fat diet (HFD) reduces sperm fertility, which is one of the cause inducing male infertility. Thus, we hypothesize that stress-induced changes in exosome content play a critical role in mediating this detrimental process. METHODS: An obese mouse model was established by feeding a HFD. Then oxidative stress status was measured in the mouse caput epididymis, epididymal fluid and spermatozoa. Meanwhile, epididymis-derived purified exosomes were isolated and validated. Subsequently, liquid chromatography tandem mass spectrometry (LC-MS) was used to perform proteomic analysis of purified exosomes. Gene Ontology (GO) analysis was performed along with pathway enrichment to identify differentially expressed proteins (DEPs). RESULTS Two hundred and two DEPs mostly related to endoplasmic reticulum (ER) function were identified in the exosomes separated from the epididymis of control mice and obese mice. The ER stress and CD63 (an exosome marker), both increased in the caput epididymis of obese mice. Furthermore, an in vitro study showed that palmitic acid (PA), an-oxidative stress inducer, increased exosome biogenesis and secretion in the EECs. CONCLUSION Oxidative stress in the epididymal microenvironment induces ER stress in the EECs. This effect alters the epididymis-derived exosome content, profile and amounts of their differentially expressed ER proteins. Such changes may affect exosome biogenesis and cargo packaging, finally leading to abnormalities in sperm maturation and fertility.
Collapse
Affiliation(s)
- Yangyang Li
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wenzhen Zhao
- Department of Histology and Embryology, School of Basic Medical Science, Dali University, 671000, Dali, Yunnan, China
| | - Rong Fu
- Department of Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Zhuoyao Ma
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yanqin Hu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yue Liu
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Zhide Ding
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Key Laboratory for Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Department of Histology, Embryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, No.280, Chongqing Road (South), 200025, Shanghai, China.
| |
Collapse
|
27
|
Vera J, Lai X, Baur A, Erdmann M, Gupta S, Guttà C, Heinzerling L, Heppt MV, Kazmierczak PM, Kunz M, Lischer C, Pützer BM, Rehm M, Ostalecki C, Retzlaff J, Witt S, Wolkenhauer O, Berking C. Melanoma 2.0. Skin cancer as a paradigm for emerging diagnostic technologies, computational modelling and artificial intelligence. Brief Bioinform 2022; 23:6761961. [PMID: 36252807 DOI: 10.1093/bib/bbac433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/28/2022] [Accepted: 09/08/2022] [Indexed: 12/19/2022] Open
Abstract
We live in an unprecedented time in oncology. We have accumulated samples and cases in cohorts larger and more complex than ever before. New technologies are available for quantifying solid or liquid samples at the molecular level. At the same time, we are now equipped with the computational power necessary to handle this enormous amount of quantitative data. Computational models are widely used helping us to substantiate and interpret data. Under the label of systems and precision medicine, we are putting all these developments together to improve and personalize the therapy of cancer. In this review, we use melanoma as a paradigm to present the successful application of these technologies but also to discuss possible future developments in patient care linked to them. Melanoma is a paradigmatic case for disruptive improvements in therapies, with a considerable number of metastatic melanoma patients benefiting from novel therapies. Nevertheless, a large proportion of patients does not respond to therapy or suffers from adverse events. Melanoma is an ideal case study to deploy advanced technologies not only due to the medical need but also to some intrinsic features of melanoma as a disease and the skin as an organ. From the perspective of data acquisition, the skin is the ideal organ due to its accessibility and suitability for many kinds of advanced imaging techniques. We put special emphasis on the necessity of computational strategies to integrate multiple sources of quantitative data describing the tumour at different scales and levels.
Collapse
Affiliation(s)
- Julio Vera
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Xin Lai
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Andreas Baur
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Michael Erdmann
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, Rostock 18051, Germany
| | - Cristiano Guttà
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Lucie Heinzerling
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany.,Department of Dermatology, LMU University Hospital, Munich, Germany
| | - Markus V Heppt
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | | | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany
| | - Christopher Lischer
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Christian Ostalecki
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Jimmy Retzlaff
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | | | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, Rostock 18051, Germany
| | - Carola Berking
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| |
Collapse
|
28
|
Muraoka S, Hirano M, Isoyama J, Ishida M, Tomonaga T, Adachi J. Automated Proteomics Sample Preparation of Phosphatidylserine-Positive Extracellular Vesicles from Human Body Fluids. ACS OMEGA 2022; 7:41472-41479. [PMID: 36406491 PMCID: PMC9670285 DOI: 10.1021/acsomega.2c05244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
Extracellular vesicles (EVs) are ubiquitously secreted by almost every cell type and are present in all body fluids. Blood-derived EVs can be used as a promising source for biomarker monitoring in disease. EV proteomics is currently being analyzed in clinical specimens. However, their EV proteomics preparation methods are limited in throughput for human subjects. Here, we introduced a novel automated EV isolation and sample preparation method using a magnetic particle processing robot for automated 96-well processing of magnetic particles for EV proteomics analysis that can be started with a low volume of multiple clinical samples. The automation of EV purification reduced the coefficient of variation of protein quantification from 3.5 to 2.2% compared with manual purification, enabling the quantification of 1120 proteins in 1 h of MS analysis. This automated proteomics EV sample preparation is attractive for processing large cohort samples for biomarker development, validation, and routine testing.
Collapse
Affiliation(s)
- Satoshi Muraoka
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Clinical and Analytical Chemistry, Collaborative Research Center
for Health and Medicine, National Institute
of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Masayo Hirano
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Junko Isoyama
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Mimiko Ishida
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
| | - Jun Adachi
- Laboratory
of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health
and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Clinical and Analytical Chemistry, Collaborative Research Center
for Health and Medicine, National Institute
of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory
of Proteomics and Drug Discovery, Graduate School of Pharmaceutical
Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
29
|
Matthiesen R, Gameiro P, Henriques A, Bodo C, Moraes MCS, Costa-Silva B, Cabeçadas J, Gomes da Silva M, Beck HC, Carvalho AS. Extracellular Vesicles in Diffuse Large B Cell Lymphoma: Characterization and Diagnostic Potential. Int J Mol Sci 2022; 23:13327. [PMID: 36362114 PMCID: PMC9654702 DOI: 10.3390/ijms232113327] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 09/29/2023] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is an aggressive B cell lymphoma characterized by a heterogeneous behavior and in need of more accurate biological characterization monitoring and prognostic tools. Extracellular vesicles are secreted by all cell types and are currently established to some extent as representatives of the cell of origin. The present study characterized and evaluated the diagnostic and prognostic potential of plasma extracellular vesicles (EVs) proteome in DLBCL by using state-of-the-art mass spectrometry. The EV proteome is strongly affected by DLBCL status, with multiple proteins uniquely identified in the plasma of DLBCL. A proof-of-concept classifier resulted in highly accurate classification with a sensitivity and specificity of 1 when tested on the holdout test data set. On the other hand, no proteins were identified to correlate with non-germinal center B-cell like (non-GCB) or GCB subtypes to a significant degree after correction for multiple testing. However, functional analysis suggested that antigen binding is regulated when comparing non-GCB and GCB. Survival analysis based on protein quantitative values and clinical parameters identified multiple EV proteins as significantly correlated to survival. In conclusion, the plasma extracellular vesicle proteome identifies DLBCL cancer patients from healthy donors and contains potential EV protein markers for prediction of survival.
Collapse
Affiliation(s)
- Rune Matthiesen
- Computational and Experimental Biology Group, NOVA Medical School-Research, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisbon, Portugal
| | - Paula Gameiro
- Instituto Português de Oncologia, Departament of Hematology, 1099-213 Lisbon, Portugal
| | - Andreia Henriques
- Computational and Experimental Biology Group, NOVA Medical School-Research, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisbon, Portugal
| | - Cristian Bodo
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | | | - Bruno Costa-Silva
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - José Cabeçadas
- Instituto Português de Oncologia, Departament of Hematology, 1099-213 Lisbon, Portugal
| | - Maria Gomes da Silva
- Instituto Português de Oncologia, Departament of Hematology, 1099-213 Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry, Odense University Hospital, DK-5000 Odense, Denmark
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, NOVA Medical School-Research, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisbon, Portugal
| |
Collapse
|
30
|
He B, Huang Z, Huang C, Nice EC. Clinical applications of plasma proteomics and peptidomics: Towards precision medicine. Proteomics Clin Appl 2022; 16:e2100097. [PMID: 35490333 DOI: 10.1002/prca.202100097] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/16/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023]
Abstract
In the context of precision medicine, disease treatment requires individualized strategies based on the underlying molecular characteristics to overcome therapeutic challenges posed by heterogeneity. For this purpose, it is essential to develop new biomarkers to diagnose, stratify, or possibly prevent diseases. Plasma is an available source of biomarkers that greatly reflects the physiological and pathological conditions of the body. An increasing number of studies are focusing on proteins and peptides, including many involving the Human Proteome Project (HPP) of the Human Proteome Organization (HUPO), and proteomics and peptidomics techniques are emerging as critical tools for developing novel precision medicine preventative measures. Excitingly, the emerging plasma proteomics and peptidomics toolbox exhibits a huge potential for studying pathogenesis of diseases (e.g., COVID-19 and cancer), identifying valuable biomarkers and improving clinical management. However, the enormous complexity and wide dynamic range of plasma proteins makes plasma proteome profiling challenging. Herein, we summarize the recent advances in plasma proteomics and peptidomics with a focus on their emerging roles in COVID-19 and cancer research, aiming to emphasize the significance of plasma proteomics and peptidomics in clinical applications and precision medicine.
Collapse
Affiliation(s)
- Bo He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China.,Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
31
|
Xue M, Ye S, Ma X, Ye F, Wang C, Zhu L, Yang Y, Chen J. Single-Vesicle Infrared Nanoscopy for Noninvasive Tumor Malignancy Diagnosis. J Am Chem Soc 2022; 144:20278-20287. [DOI: 10.1021/jacs.2c07393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mengfei Xue
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyuan Ye
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Xiaopeng Ma
- The First Affiliated Hospital University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui 230000, China
| | - Fangfu Ye
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Oujiang Laboratory, Wenzhou, Zhejiang 325000, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianing Chen
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| |
Collapse
|
32
|
Extracellular vesicle isolation, purification and evaluation in cancer diagnosis. Expert Rev Mol Med 2022; 24:e41. [PMID: 36268744 DOI: 10.1017/erm.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Strategies for non-invasive biomarker discovery in early detection of cancer are an urgent need. Extracellular vesicles (EVs) have generated increasing attention from the scientific community and are under intensive investigations due to their unique biological profiles and their non-invasive nature. EVs are membrane-enclosed vesicles with variable sizes and function. Such vesicles are actively secreted from multiple cell types and are considered as key vehicles for inter-cellular communications and signalling. The stability and potential to easily cross biological barriers enable EVs for exerting durable effects on target cells. These along with easy access to such vesicles, the consistent secretion from tumour during all stages of tumorigenesis and their content providing a reservoir of molecules as well as mirroring the identity of the cell of origin are virtues that have made EVs appealing to be assessed in liquid biopsy approaches and for using as a promising resource of biomarkers in cancer diagnosis and therapy and monitoring targeted cancer therapy. Early detection of EVs will guide time-scheduled personalised therapy. Surveying reliable and sensitive methods for rapid isolation of EVs from biofluids, the purity of isolated vesicles and their molecular profiling and marker specification for clinical translation in patients with cancer are issues in the area and the hot topics of many recent studies. Here, the focus is over methods for EV isolation and stratification for digging more information about liquid biopsy-based diagnosis. Extending knowledge regarding EV-based strategies is a key to validate independent patient follow-up for cancer diagnosis at early stages and inspecting the efficacy of therapeutics.
Collapse
|
33
|
The Role of Extracellular Vesicles in Melanoma Progression. Cancers (Basel) 2022; 14:cancers14133086. [PMID: 35804857 PMCID: PMC9264817 DOI: 10.3390/cancers14133086] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 02/07/2023] Open
Abstract
Cutaneous melanoma arises from a malignant transformation of the melanocytes in the skin. It is the deadliest form of skin cancer owing to its potential to metastasize. While recent advances in immuno-oncology have been successful in melanoma treatment, not all the patients respond to the treatment equally, thus individual pre-screening and personalized combination therapies are essential to stratify and monitor patients. Extracellular vesicles (EVs) have emerged as promising biomarker candidates to tackle these challenges. EVs are ~50-1000-nm-sized, lipid bilayer-enclosed spheres, which are secreted by almost all cell types, including cancer cells. Their cargo, such as nucleic acids, proteins, lipids, amino acids, and metabolites, can be transferred to target cells. Thanks to these properties, EVs can both provide a multiplexed molecular fingerprint of the cell of origin and thus serve as potential biomarkers, or reveal pathways important for cancer progression that can be targeted pharmaceutically. In this review we give a general overview of EVs and focus on their impact on melanoma progression. In particular, we shed light on the role of EVs in shaping the tumor-stroma interactions that facilitate metastasis and summarize the latest findings on molecular profiling of EV-derived miRNAs and proteins that can serve as potential biomarkers for melanoma progression.
Collapse
|
34
|
Zebrowska A, Jelonek K, Mondal S, Gawin M, Mrowiec K, Widłak P, Whiteside T, Pietrowska M. Proteomic and Metabolomic Profiles of T Cell-Derived Exosomes Isolated from Human Plasma. Cells 2022; 11:1965. [PMID: 35741093 PMCID: PMC9222142 DOI: 10.3390/cells11121965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 02/04/2023] Open
Abstract
Exosomes that are released by T cells are key messengers involved in immune regulation. However, the molecular profiling of these vesicles, which is necessary for understanding their functions, requires their isolation from a very heterogeneous mixture of extracellular vesicles that are present in the human plasma. It has been shown that exosomes that are produced by T cells could be isolated from plasma by immune capture using antibodies that target the CD3 antigen, which is a key component of the TCR complex that is present in all T lymphocytes. Here, we demonstrate that CD3(+) exosomes that are isolated from plasma can be used for high-throughput molecular profiling using proteomics and metabolomics tools. This profiling allowed for the identification of proteins and metabolites that differentiated the CD3(+) from the CD3(-) exosome fractions that were present in the plasma of healthy donors. Importantly, the proteins and metabolites that accumulated in the CD3(+) vesicles reflected the known molecular features of T lymphocytes. Hence, CD3(+) exosomes that are isolated from human plasma by immune capture could serve as a "T cell biopsy".
Collapse
Affiliation(s)
- Aneta Zebrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland; (A.Z.); (K.J.); (M.G.); (K.M.)
| | - Karol Jelonek
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland; (A.Z.); (K.J.); (M.G.); (K.M.)
| | - Sujan Mondal
- UPMC Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA;
| | - Marta Gawin
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland; (A.Z.); (K.J.); (M.G.); (K.M.)
| | - Katarzyna Mrowiec
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland; (A.Z.); (K.J.); (M.G.); (K.M.)
| | - Piotr Widłak
- Clinical Research Support Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Theresa Whiteside
- UPMC Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA;
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Monika Pietrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland; (A.Z.); (K.J.); (M.G.); (K.M.)
| |
Collapse
|
35
|
Proteomic and Biochemical Analysis of Extracellular Vesicles Isolated from Blood Serum of Patients with Melanoma. SEPARATIONS 2022. [DOI: 10.3390/separations9040086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background: Malignant melanoma is the most serious type of skin cancer with the highest mortality rate. Extracellular vesicles (EVs) have potential as new tumor markers that could be used as diagnostic and prognostic markers for early detection of melanoma. Methods: EVs were purified from the blood serum of melanoma patients using two methods—ultracentrifugation and PEG precipitation—and analyzed by mass spectrometry and immunoblot. Results: We identified a total of 585 unique proteins; 334 proteins were detected in PEG-precipitated samples and 515 in UC-purified EVs. EVs purified from patients varied in their size and concentration in different individuals. EVs obtained from stage II and III patients were, on average, smaller and more abundant than others. Detailed analysis of three potential biomarkers—SERPINA3, LGALS3BP, and gelsolin—revealed that the expression of SERPINA3 and LGALS3BP was higher in melanoma patients than healthy controls, while gelsolin exhibited higher expression in healthy controls. Conclusion: We suggest that all three proteins might have potential to be used as biomarkers, but a number of issues, such as purification of EVs, standardization, and validation of methods suitable for everyday clinical settings, still need to be addressed.
Collapse
|
36
|
Sun K, Pan L, Chen J, Deng Y, Chen J, Tian Z, Tang GY, Wang Y. Protein disulfide isomerase family A member 3 expression is upregulated in tissue-derived extracellular vesicles in oral lichen planus and oral lichenoid lesions. Arch Oral Biol 2022; 137:105390. [DOI: 10.1016/j.archoralbio.2022.105390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/02/2022]
|
37
|
Campos-Silva C, Cáceres-Martell Y, Sánchez-Herrero E, Sandúa A, Beneitez-Martínez A, González Á, Provencio M, Romero A, Jara-Acevedo R, Yáñez-Mó M, Valés-Gómez M. A simple immunoassay for extracellular vesicle liquid biopsy in microliters of non-processed plasma. J Nanobiotechnology 2022; 20:72. [PMID: 35135541 PMCID: PMC8822649 DOI: 10.1186/s12951-022-01256-5] [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: 10/18/2021] [Accepted: 01/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs), released by most cell types, provide an excellent source of biomarkers in biological fluids. However, in order to perform validation studies and screenings of patient samples, it is still necessary to develop general techniques permitting rapid handling of small amounts of biological samples from large numbers of donors. RESULTS Here we describe a method that, using just a few microliters of patient's plasma, identifies tumour markers exposed on EVs. Studying physico-chemical properties of EVs in solution, we demonstrate that they behave as stable colloidal suspensions and therefore, in immunocapture assays, many of them are unable to interact with a stationary functionalised surface. Using flocculation methods, like those used to destabilize colloids, we demonstrate that cationic polymers increase EV ζ-potential, diameter, and sedimentation coefficient and thus, allow a more efficient capture on antibody-coated surfaces by both ELISA and bead-assisted flow cytometry. These findings led to optimization of a protocol in microtiter plates allowing effective immunocapture of EVs, directly in plasma without previous ultracentrifugation or other EV enrichment. The method, easily adaptable to any laboratory, has been validated using plasma from lung cancer patients in which the epithelial cell marker EpCAM has been detected on EVs. CONCLUSIONS This optimized high throughput, easy to automate, technology allows screening of large numbers of patients to phenotype tumour markers in circulating EVs, breaking barriers for the validation of proposed EV biomarkers and the discovery of new ones.
Collapse
Affiliation(s)
- Carmen Campos-Silva
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Yaiza Cáceres-Martell
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Estela Sánchez-Herrero
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Atrys Health, Barcelona, Spain
| | - Amaia Sandúa
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Álvaro González
- Service of Biochemistry, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Provencio
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Atocha Romero
- Laboratorio de Biopsia Líquida, Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | | | - María Yáñez-Mó
- Department of Molecular Biology, UAM - Centro de Biología Molecular Severo Ochoa, Madrid, Spain.,Instituto de Investigación del Hospital Universitario La Princesa, Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology, CNB-CSIC, Madrid, Spain.
| |
Collapse
|
38
|
Bao Q, Huang Q, Chen Y, Wang Q, Sang R, Wang L, Xie Y, Chen W. Tumor-Derived Extracellular Vesicles Regulate Cancer Progression in the Tumor Microenvironment. Front Mol Biosci 2022; 8:796385. [PMID: 35059436 PMCID: PMC8764126 DOI: 10.3389/fmolb.2021.796385] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized particles released by numerous kinds of cells, which are now increasingly considered as essential vehicles of cell-to-cell communication and biomarkers in disease diagnosis and treatment. They contain a variety of biomolecular components, including lipids, proteins and nucleic acids. These functional molecules can be transmitted between tumor cells and other stromal cells such as endothelial cells, fibroblasts and immune cells utilizing EVs. As a result, tumor-derived EVs can deliver molecules to remodel the tumor microenvironment, thereby influencing cancer progression. On the one hand, tumor-derived EVs reprogram functions of endothelial cells, promote cancer-associated fibroblasts transformation, induce resistance to therapy and inhibit the immune response to form a pro-tumorigenic environment. On the other hand, tumor-derived EVs stimulate the immune response to create an anti-tumoral environment. This article focuses on presenting a comprehensive and critical overview of the potential role of tumor-derived EVs-mediated communication in the tumor microenvironment.
Collapse
Affiliation(s)
- Qianqian Bao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Qianqian Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yunna Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Qiang Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ran Sang
- Bengbu Medical College, Bengbu, China.,The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ying Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Weidong Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
39
|
Boussadia Z, Gambardella AR, Mattei F, Parolini I. Acidic and Hypoxic Microenvironment in Melanoma: Impact of Tumour Exosomes on Disease Progression. Cells 2021; 10:3311. [PMID: 34943819 PMCID: PMC8699343 DOI: 10.3390/cells10123311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of melanoma progression have been extensively studied in the last decade, and despite the diagnostic and therapeutic advancements pursued, malignant melanoma still accounts for 60% of skin cancer deaths. Therefore, research efforts are required to better define the intercellular molecular steps underlying the melanoma development. In an attempt to represent the complexity of the tumour microenvironment (TME), here we analysed the studies on melanoma in acidic and hypoxic microenvironments and the interactions with stromal and immune cells. Within TME, acidity and hypoxia force melanoma cells to adapt and to evolve into a malignant phenotype, through the cooperation of the tumour-surrounding stromal cells and the escape from the immune surveillance. The role of tumour exosomes in the intercellular crosstalk has been generally addressed, but less studied in acidic and hypoxic conditions. Thus, this review aims to summarize the role of acidic and hypoxic microenvironment in melanoma biology, as well as the role played by melanoma-derived exosomes (Mexo) under these conditions. We also present a perspective on the characteristics of acidic and hypoxic exosomes to disclose molecules, to be further considered as promising biomarkers for an early detection of the disease. An update on the use of exosomes in melanoma diagnosis, prognosis and response to treatment will be also provided and discussed.
Collapse
Affiliation(s)
- Zaira Boussadia
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Adriana Rosa Gambardella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Isabella Parolini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
40
|
Mondal SK, Whiteside TL. Proteomic profiles of melanoma cell-derived exosomes in plasma: discovery of potential biomarkers of melanoma progression. Melanoma Res 2021; 31:472-475. [PMID: 34284459 PMCID: PMC8429197 DOI: 10.1097/cmr.0000000000000762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cancer liquid biopsy encompassing circulating tumor cells (CTC), circulating tumor DNA (ctDNA) and/or tumor-derived exosomes (TEX) emerges as a novel approach to early detection, noninvasive monitoring of responses to therapy and predicting patient survival. TEX are a key component of liquid biopsy because they mimic tumor cells in their proteomic and genetic content. Two recent proteomic analyses of TEX released into plasma by melanoma cells confirms the potential of TEX as diagnostic and prognostic markers in melanoma.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Theresa L. Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
41
|
The Fatty Acid and Protein Profiles of Circulating CD81-Positive Small Extracellular Vesicles Are Associated with Disease Stage in Melanoma Patients. Cancers (Basel) 2021; 13:cancers13164157. [PMID: 34439311 PMCID: PMC8392159 DOI: 10.3390/cancers13164157] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Early detection of cutaneous melanoma is the key to increasing survival and proper therapeutic adjustment, especially in stages II–IV. We investigated whether the fatty acid (FA) and protein compositions of small extracellular vesicles (sEV) expressing CD81, derived from the plasma of stage 0–I, II and III–IV melanoma patients, could reflect disease stage. Results showed a higher content of FA and differences in C18:0/C18:1 ratio, a marker of cell membrane fluidity, that distinguished patients’ CD81sEV from those of healthy donors (HD). By proteomic analysis (identifier PXD024434) we identified significant increases in CD14, PON1, PON3 and APOA5 in stage II CD81sEV compared to HD. In stage III–IV, CD81sEV’ RAP1B expression was decreased. These stage-related signatures may support the potential of sEV to provide information for early diagnosis, prediction of metastatic behavior, treatment and follow-up of melanoma patients. Abstract The early detection of cutaneous melanoma, a potentially lethal cancer with rising incidence, is fundamental to increasing survival and therapeutic adjustment. In stages II–IV especially, additional indications for adjuvant therapy purposes after resection and for treatment of metastatic patients are urgently needed. We investigated whether the fatty acid (FA) and protein compositions of small extracellular vesicles (sEV) derived from the plasma of stage 0–I, II and III–IV melanoma patients (n = 38) could reflect disease stage. The subpopulation of sEV expressing CD81 EV marker (CD81sEV) was captured by an ad hoc immune affinity technique from plasma depleted of large EV. Biological macromolecules were investigated by gas chromatography and mass spectrometry in CD81sEV. A higher content of FA was detectable in patients with respect to healthy donors (HD). Moreover, a higher C18:0/C18:1 ratio, as a marker of cell membrane fluidity, distinguished early (stage 0–I) from late (III–IV) stages’ CD81sEV. Proteomics detected increases in CD14, PON1, PON3 and APOA5 exclusively in stage II CD81sEV, and RAP1B was decreased in stage III–IV CD81sEV, in comparison to HD. Our results suggest that stage dependent alterations in CD81sEV’ FA and protein composition may occur early after disease onset, strengthening the potential of circulating sEV as a source of discriminatory information for early diagnosis, prediction of metastatic behavior and following up of melanoma patients.
Collapse
|
42
|
Hu C, Zhang Y, Zhang M, Li T, Zheng X, Guo Q, Zhang X. Exosomal Cripto-1 Serves as a Potential Biomarker for Perihilar Cholangiocarcinoma. Front Oncol 2021; 11:730615. [PMID: 34434900 PMCID: PMC8380828 DOI: 10.3389/fonc.2021.730615] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Perihilar cholangiocarcinoma (PHCCA) has a poor prognosis, mainly due to diagnosis at an advanced stage. Cripto-1 functions as an oncogene and is highly expressed in several human cancers, however, its clinical application in PHCCA is poorly understood. Herein, we identified that Cripto-1 was released by PHCCA cells via exosomes in vitro and in vivo. Furthermore, an ELISA method was developed to detect exosomal Cripto-1 in the serum of 115 PHCCA patients, 47 cholangitis patients and 65 healthy controls, and it was found that exosomal Cripto-1 was increased in PHCCA patients and associated with metastasis. Compared with traditional serum tumor markers, CA19-9 and CEA, exosomal Cripto-1 demonstrated a larger area under ROC curve for PHCCA diagnosis. The cutoff value of exosomal Cripto-1 was 0.82, achieving a sensitivity of 79.1% and a specificity of 87.5%. As expected, exosomal Cripto-1 levels in immunohistochemically Cripto-1-high cases were significantly elevated compared to in Cripto-1-low cases. When measured 1-week postoperatively, Cripto-1 levels decreased on average from 1.25(0.96-3.26) to 0.85(0.62-1.82). Immunohistochemistry analysis showed Cripto-1 expression was negatively correlated with E-cadherin and was an independent prognostic biomarker for poor survival in PHCCA patients. In conclusion, exosomal Cripto-1 in sera can reflect its expression in the tissue of PHCAA patients and has the potential be a non-invasive biomarker for diagnosis and prognosis of PHCCA.
Collapse
Affiliation(s)
- Chunxiao Hu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yanli Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, China
| | - Mengjiao Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Tingting Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zheng
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, China
| | - Qining Guo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
43
|
Pietrowska M, Zebrowska A, Gawin M, Marczak L, Sharma P, Mondal S, Mika J, Polańska J, Ferrone S, Kirkwood JM, Widlak P, Whiteside TL. Proteomic profile of melanoma cell-derived small extracellular vesicles in patients' plasma: a potential correlate of melanoma progression. J Extracell Vesicles 2021; 10:e12063. [PMID: 33613873 PMCID: PMC7876545 DOI: 10.1002/jev2.12063] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/07/2020] [Accepted: 01/12/2021] [Indexed: 02/04/2023] Open
Abstract
Molecular profiling of small extracellular vesicles (sEV) isolated from plasma of cancer patients emerges as promising strategy for biomarkers discovery. We investigated the proteomic profiles of sEV immunoselected using anti-CSPG4 antibodies from 15 melanoma patients' plasma. The proteomes of sEV separated into melanoma cell-derived (MTEX) and non-malignant cell-derived (NMTEX) were compared using high-resolution mass spectrometry. Paired analysis identified the MTEX-associated profile of 16 proteins that discriminated MTEX from NMETEX. We also identified the MTEX profile that discriminated between seven patients with no evidence of melanoma (NED) after therapy and eight with progressive disease (PD). Among 75 MTEX proteins overexpressed in PD patients, PDCD6IP (ALIX) had the highest discriminating value, while CNTN1 (contactin-1) was upregulated only in MTEX of NED patients. This is the first report documenting that proteomes of tumour-derived sEV in patients' plasma discriminate cancer from non-cancer and identify proteins with potential to serve as prognostic biomarkers in melanoma.
Collapse
Affiliation(s)
- Monika Pietrowska
- Maria Sklodowska‐Curie National Research Institute of OncologyGliwicePoland
| | - Aneta Zebrowska
- Maria Sklodowska‐Curie National Research Institute of OncologyGliwicePoland
| | - Marta Gawin
- Maria Sklodowska‐Curie National Research Institute of OncologyGliwicePoland
| | - Lukasz Marczak
- European Center for Bioinformatics and GenomicsInstitute of Bioorganic Chemistry PASPoznanPoland
| | - Priyanka Sharma
- UPMC Hillman Cancer CenterUniversity of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA
| | - Sujan Mondal
- UPMC Hillman Cancer CenterUniversity of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA
| | - Justyna Mika
- Department of Data Science and Engineering, Silesian University of TechnologyGliwicePoland
| | - Joanna Polańska
- Department of Data Science and Engineering, Silesian University of TechnologyGliwicePoland
| | - Soldano Ferrone
- Department of SurgeryHarvard Medical School, Massachusetts General HospitalBostonMassachusettsUSA
| | - John M. Kirkwood
- UPMC Hillman Cancer CenterUniversity of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA,Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Piotr Widlak
- Maria Sklodowska‐Curie National Research Institute of OncologyGliwicePoland
| | - Theresa L. Whiteside
- UPMC Hillman Cancer CenterUniversity of Pittsburgh Cancer InstitutePittsburghPennsylvaniaUSA,Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|