1
|
Batista IA, Machado JC, Melo SA. Advances in exosomes utilization for clinical applications in cancer. Trends Cancer 2024; 10:947-968. [PMID: 39168775 DOI: 10.1016/j.trecan.2024.07.010] [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: 03/28/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Exosomes are regarded as having transformative potential for clinical applications. Exosome-based liquid biopsies offer a noninvasive method for early cancer detection and real-time disease monitoring. Clinical trials are underway to validate the efficacy of exosomal biomarkers for enhancing diagnostic accuracy and predicting treatment responses. Additionally, engineered exosomes are being developed as targeted drug delivery systems that can navigate the bloodstream to deliver therapeutic agents to tumor sites, thus enhancing treatment efficacy while minimizing systemic toxicity. Exosomes also exhibit immunomodulatory properties, which are being harnessed to boost antitumor immune responses. In this review, we detail the latest advances in clinical trials and research studies, underscoring the potential of exosomes to revolutionize cancer care.
Collapse
Affiliation(s)
- Inês A Batista
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - José C Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; P.CCC Porto Comprehensive Cancer Centre, Raquel Seruca, Portugal
| | - Sonia A Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; P.CCC Porto Comprehensive Cancer Centre, Raquel Seruca, Portugal.
| |
Collapse
|
2
|
Kalvala AK, Silwal A, Patel B, Kasetti A, Shetty K, Cho JH, Lara G, Daugherity B, Diesler R, Pooladanda V, Rueda BR, Henske EP, Yu JJ, Markiewski M, Karbowniczek M. Extracellular vesicles regulate metastable phenotypes of lymphangioleiomyomatosis cells via shuttling ATP synthesis to pseudopodia and activation of integrin adhesion complexes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.611297. [PMID: 39314494 PMCID: PMC11419057 DOI: 10.1101/2024.09.09.611297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is metastatic sarcoma but mechanisms regulating LAM metastasis are unknown. Extracellular vesicle (EV) regulate cancer metastasis but their roles in LAM have not yet been investigated. Here, we report that EV biogenesis is increased in LAM and LAM EV cargo is enriched with lung tropic integrins, metalloproteinases, and cancer stem cell markers. LAM-EV increase LAM cell migration and invasion via the ITGα6/β1-c-Src-FAK-AKT axis. Metastable (hybrid) phenotypes of LAM metastasizing cells, pivotal for metastasis, are regulated by EV from primary tumor or metastasizing LAM cells via shuttling ATP synthesis to cell pseudopodia or activation of integrin adhesion complex, respectively. In mouse models of LAM, LAM-EV increase lung metastatic burden through mechanisms involving lung extracellular matrix remodeling. Collectively, these data provide evidence for the role of EV in promoting LAM lung metastasis and identify novel EV-dependent mechanisms regulating metastable phenotypes of tumor cells. Clinical impact of research is that it establishes LAM pathway as novel target for LAM therapy.
Collapse
|
3
|
Fattahi AS, Jafari M, Farahavar G, Abolmaali SS, Tamaddon AM. Expanding horizons in cancer therapy by immunoconjugates targeting tumor microenvironments. Crit Rev Oncol Hematol 2024; 201:104437. [PMID: 38977144 DOI: 10.1016/j.critrevonc.2024.104437] [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: 03/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Immunoconjugates are promising molecules combining antibodies with different agents, such as toxins, drugs, radionuclides, or cytokines that primarily aim to target tumor cells. However, tumor microenvironment (TME), which comprises a complex network of various cells and molecular cues guiding tumor growth and progression, remains a major challenge for effective cancer therapy. Our review underscores the pivotal role of TME in cancer therapy with immunoconjugates, examining the intricate interactions with TME and recent advancements in TME-targeted immunoconjugates. We explore strategies for targeting TME components, utilizing diverse antibodies such as neutralizing, immunomodulatory, immune checkpoint inhibitors, immunostimulatory, and bispecific antibodies. Additionally, we discuss different immunoconjugates, elucidating their mechanisms of action, advantages, limitations, and applications in cancer immunotherapy. Furthermore, we highlight emerging technologies enhancing the safety and efficacy of immunoconjugates, such as antibody engineering, combination therapies, and nanotechnology. Finally, we summarize current advancements, perspectives, and future developments of TME-targeted immunoconjugates.
Collapse
Affiliation(s)
- Amir Saamaan Fattahi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| |
Collapse
|
4
|
Poinsot V, Pizzinat N, Ong-Meang V. Engineered and Mimicked Extracellular Nanovesicles for Therapeutic Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:639. [PMID: 38607173 PMCID: PMC11013861 DOI: 10.3390/nano14070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Exosomes are spherical extracellular nanovesicles with an endosomal origin and unilamellar lipid-bilayer structure with sizes ranging from 30 to 100 nm. They contain a large range of proteins, lipids, and nucleic acid species, depending on the state and origin of the extracellular vesicle (EV)-secreting cell. EVs' function is to encapsulate part of the EV-producing cell content, to transport it through biological fluids to a targeted recipient, and to deliver their cargos specifically within the aimed recipient cells. Therefore, exosomes are considered to be potential biological drug-delivery systems that can stably deliver their cargo into targeted cells. Various cell-derived exosomes are produced for medical issues, but their use for therapeutic purposes still faces several problems. Some of these difficulties can be avoided by resorting to hemisynthetic approaches. We highlight here the uses of alternative exosome-mimes involving cell-membrane coatings on artificial nanocarriers or the hybridization between exosomes and liposomes. We also detail the drug-loading strategies deployed to make them drug-carrier systems and summarize the ongoing clinical trials involving exosomes or exosome-like structures. Finally, we summarize the open questions before considering exosome-like disposals for confident therapeutic delivery.
Collapse
Affiliation(s)
- Verena Poinsot
- Inserm, CNRS, Faculté de Santé, Université Toulouse III—Paul Sabatier, I2MC U1297, 31432 Toulouse, France; (N.P.); (V.O.-M.)
| | | | | |
Collapse
|
5
|
Cai X, Wang Z, Lin S, Chen H, Bu H. Ginsenoside Rg3 suppresses vasculogenic mimicry by impairing DVL3-maintained stemness via PAAD cell-derived exosomal miR-204 in pancreatic adenocarcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155402. [PMID: 38350242 DOI: 10.1016/j.phymed.2024.155402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Vasculogenic mimicry (VM) is an angiogenesis-independent process that potentially contributes to the poor clinical outcome of anti-angiogenesis therapy in multiple malignant cancers, including pancreatic adenocarcinoma (PAAD). Several studies have shown that ginsenoside Rg3, a bioactive component of ginseng, holds considerable potential for cancer treatment. Our previous work has proved that Rg3 can inhibit VM formation in PAAD. However, its underlying mechanism remains unclear. PURPOSE To explore the underlying mechanism by which Rg3 affects VM formation in PAAD. METHODS We first investigated the effects of Rg3 on the cellular phenotypes of two PAAD cell lines (SW-1990 and PCI-35), and the expression of EMT- and stemness-related proteins. SW-1990 cells were adopted to construct xenograft models, and the anti-tumor effects of Rg3 in vivo were validated. Subsequently, we isolated the exosomes from the two PAAD cell lines with Rg3 treatment or not, and explored whether Rg3 regulated VM via PAAD cell-derived exosomes. MiRNA sequencing, clinical analysis, and rescue experiments were performed to investigate whether and which miRNA was involved. Subsequently, the target gene of miRNA was predicted using the miRDB website (https://mirdb.org/), and rescue experiments were further conducted to validate those in vitro and in vivo. RESULTS Rg3 indeed exhibited excellent anti-tumor effects both in vitro and in vivo, with inhibitory effects on EMT and stemness of PAAD cells. More interestingly, Rg3-treated PAAD cell-derived exosomes suppressed the tube-forming ability of HUVEC and PAAD cells, with a decrease in stemness-related protein expression, indicating that Rg3 inhibited both angiogenesis and VM processes. Subsequently, we found that Rg3 induced the up-regulation of miR-204 in PAAD cell-derived exosomes, and miR-204 alone inhibited tube and sphere formation abilities of PAAD cells like exosomes. Specifically, miR-204 down-regulated DVL3 expression, which was involved in regulating cancer cell stemness, and ultimately affected VM. The in vivo experiments further indicated that Rg3-treated SW-1990 cell-derived exosome-inhibited tumor growth, VM formation, and stemness-related protein expression can be abrogated by DVL3 overexpression. CONCLUSION Ginsenoside Rg3 increased the PAAD cell-derived exosomal miR-204 levels, which subsequently inhibited its target genes DVL3 expression in the receptor PAAD cells, and the down-regulated DVL3 broke stemness maintenance, ultimately suppressing VM formation of PAAD. Our findings revealed a novel mechanism by which Rg3 exerted its anti-tumor activity in PAAD via inhibiting VM, and provided a promising strategy to make up for the deficiency of anti-angiogenesis therapy in cancer.
Collapse
Affiliation(s)
- Xufan Cai
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China
| | - Zhaohong Wang
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Shengzhang Lin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, #51 Huzhou Street, Gongshu District, Hangzhou, Zhejiang 310015, People's Republic of China.
| | - Hui Chen
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Heqi Bu
- Department of Surgery, Tongde Hospital of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310012, People's Republic of China
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [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
|
7
|
Chak PT, Kam NW, Choi TH, Dai W, Kwong DLW. Unfolding the Complexity of Exosome-Cellular Interactions on Tumour Immunity and Their Clinical Prospects in Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:919. [PMID: 38473281 DOI: 10.3390/cancers16050919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy situated in the posterolateral nasopharynx. NPC poses grave concerns in Southeast Asia due to its late diagnosis. Together with resistance to standard treatment combining chemo- and radiotherapy, NPC presents high metastatic rates and common recurrence. Despite advancements in immune-checkpoint inhibitors (ICIs) and cytotoxic-T-lymphocytes (CTLs)-based cellular therapy, the exhaustive T cell profile and other signs of immunosuppression within the NPC tumour microenvironment (TME) remain as concerns to immunotherapy response. Exosomes, extracellular vesicles of 30-150 nm in diameter, are increasingly studied and linked to tumourigenesis in oncology. These bilipid-membrane-bound vesicles are packaged with a variety of signalling molecules, mediating cell-cell communications. Within the TME, exosomes can originate from tumour, immune, or stromal cells. Although there are studies on tumour-derived exosomes (TEX) in NPC and their effects on tumour processes like angiogenesis, metastasis, therapeutic resistance, there is a lack of research on their involvement in immune evasion. In this review, we aim to enhance the comprehension of how NPC TEX contribute to cellular immunosuppression. Furthermore, considering the detectability of TEX in bodily fluids, we will also discuss the potential development of TEX-related biomarkers for liquid biopsy in NPC as this could facilitate early diagnosis and prognostication of the disease.
Collapse
Affiliation(s)
- Paak-Ting Chak
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Tsz-Ho Choi
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
8
|
Adem B, Bastos N, Ruivo CF, Sousa-Alves S, Dias C, Vieira PF, Batista IA, Cavadas B, Saur D, Machado JC, Cai D, Melo SA. Exosomes define a local and systemic communication network in healthy pancreas and pancreatic ductal adenocarcinoma. Nat Commun 2024; 15:1496. [PMID: 38383468 PMCID: PMC10881969 DOI: 10.1038/s41467-024-45753-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/04/2024] [Indexed: 02/23/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a lethal disease, requires a grasp of its biology for effective therapies. Exosomes, implicated in cancer, are poorly understood in living systems. Here we use the genetically engineered mouse model (ExoBow) to map the spatiotemporal distribution of exosomes from healthy and PDAC pancreas in vivo to determine their biological significance. We show that, within the PDAC microenvironment, cancer cells establish preferential communication routes through exosomes with cancer associated fibroblasts and endothelial cells. The latter being a conserved event in the healthy pancreas. Inhibiting exosomes secretion in both scenarios enhances angiogenesis, underscoring their contribution to vascularization and to cancer. Inter-organ communication is significantly increased in PDAC with specific organs as most frequent targets of exosomes communication occurring in health with the thymus, bone-marrow, brain, and intestines, and in PDAC with the kidneys, lungs and thymus. In sum, we find that exosomes mediate an organized intra- and inter- pancreas communication network with modulatory effects in vivo.
Collapse
Affiliation(s)
- Bárbara Adem
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Nuno Bastos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Carolina F Ruivo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Sara Sousa-Alves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Carolina Dias
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Patrícia F Vieira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Inês A Batista
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Cavadas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Dieter Saur
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - José C Machado
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- P.CCC Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal
| | - Dawen Cai
- Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
- Biophysics, LS&A, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sonia A Melo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
- P.CCC Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal.
| |
Collapse
|
9
|
Zhao Z, Yang S, Tang X, Feng L, Ding Z, Chen Z, Luo X, Deng R, Sheng J, Xie S, Chang K, Chen M. DNA four-way junction-driven dual-rolling circle amplification sandwich-type aptasensor for ultra-sensitive and specific detection of tumor-derived exosomes. Biosens Bioelectron 2024; 246:115841. [PMID: 38006701 DOI: 10.1016/j.bios.2023.115841] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/27/2023]
Abstract
There is an urgent need to accurately quantify tumor-derived exosomes, which have emerged as promising non-invasive tumor diagnostic biomarkers. Herein, a bispecific-aptamer sandwich-type gold nanoparticle-modified electrochemical aptasensor was developed based on a four-way junction (4-WJ)-triggered dual rolling circle amplification (RCA)-assisted methylene blue (MB)/G-quadruplex strategy for extremely specific and sensitive exosome detection. This aptamer/exosome/aptamer sandwich-type design contained a CD63-specific aptamer and a cancerous mucin-1 (MUC1) protein-specific aptamer. The CD63 aptamer modified on a gold electrode captured exosomes, and then the sandwich-type aptasensor was formed with the addition of the MUC1 aptamer. The MUC1 aptamer's 3'-end sequence facilitated the formation of 4-WJ, assisted by a molecular beacon probe and a binary DNA probe. Subsequently, a dual-RCA reaction was triggered by binding to two cytosine-rich circle DNA templates at both ends of 4-WJ. Ultimately, dual-RCA products containing multiple G-quadruplex conformations were generated with the assistance of K+ to trap abundant MB indicators and amplify electrochemical signals. The aptasensor exhibited high specificity, sensitivity, repeatability, and stability toward MCF-7-derived exosomes, with a detection limit of 20 particles/mL and a linear range of 1 × 102 to 1 × 107 particles/mL. Moreover, it showed excellent applicability in clinical settings to recover exosomes in normal human serum. Our aptasensor is anticipated to serve as a versatile platform for detecting various specific aptamer-based targets in biomedical and bioanalytical applications.
Collapse
Affiliation(s)
- Zhuyang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Sha Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Liu Feng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Zishan Ding
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Zhiguo Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xing Luo
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Ruijia Deng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Jing Sheng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Shuang Xie
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China; College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
10
|
Chen W, Zhou M, Guan B, Xie B, Liu Y, He J, Zhao J, Zhao Q, Yan D. Tumour-associated macrophage-derived DOCK7-enriched extracellular vesicles drive tumour metastasis in colorectal cancer via the RAC1/ABCA1 axis. Clin Transl Med 2024; 14:e1591. [PMID: 38385857 PMCID: PMC10883245 DOI: 10.1002/ctm2.1591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/28/2024] [Accepted: 02/03/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Metastasis accounts for the majority of deaths among patients with colorectal cancer (CRC). Here, the regulatory role of tumour-associated macrophages (TAMs) in CRC metastasis was explored. METHODS Immunohistochemical (IHC) analysis of the TAM biomarker CD163 was conducted to evaluate TAM infiltration in CRC. Transwell assays and an ectopic liver metastasis model were established to evaluate the metastatic ability of tumour cells. RNA sequencing (RNA-seq) and liquid chromatography-mass spectrometry (LC-MS) were applied to identify the differentially expressed genes and proteins in CRC cells and in TAM-derived extracellular vesicles (EVs). Cholesterol content measurement, a membrane fluidity assay and filipin staining were performed to evaluate cholesterol efflux in CRC cells. RESULTS Our results showed that TAM infiltration is positively correlated with CRC metastasis. TAMs can facilitate the migration and invasion of MC-38 and CT-26 cells via EVs. According to the RNA-seq data, TAM-EVs increase cholesterol efflux and enhance membrane fluidity in CRC cells by regulating ABCA1 expression, thus affecting the motility of CRC cells. Mechanistically, DOCK7 packaged in TAM-EVs can activate RAC1 in CRC cells and subsequently upregulate ABCA1 expression by phosphorylating AKT and FOXO1. Moreover, IHC analysis of ABCA1 in patients with liver-metastatic CRC indicated that ABCA1 expression is significantly greater in metastatic liver nodules than in primary CRC tumours. CONCLUSIONS Overall, our findings suggest that DOCK7 delivered via TAM-EVs could regulate cholesterol metabolism in CRC cells and CRC cell metastasis through the RAC1/AKT/FOXO1/ABCA1 axis. DOCK7 could thus be a new therapeutic target for controlling CRC metastasis.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Menghua Zhou
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bingjie Guan
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bowen Xie
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Youdong Liu
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiang He
- Department of General SurgeryDongTai People's HospitalDongtaiJiangsuChina
| | - Jingjing Zhao
- Department of General SurgeryDongTai People's HospitalDongtaiJiangsuChina
| | - Qian Zhao
- Department of PathophysiologyKey Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dongwang Yan
- Department of General SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Xiong Y, Lou P, Xu C, Han B, Liu J, Gao J. Emerging role of extracellular vesicles in veterinary practice: novel opportunities and potential challenges. Front Vet Sci 2024; 11:1335107. [PMID: 38332755 PMCID: PMC10850357 DOI: 10.3389/fvets.2024.1335107] [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: 11/08/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Extracellular vesicles are nanoscale vesicles that transport signals between cells, mediating both physiological and pathological processes. EVs facilitate conserved intercellular communication. By transferring bioactive molecules between cells, EVs coordinate systemic responses, regulating homeostasis, immunity, and disease progression. Given their biological importance and involvement in pathogenesis, EVs show promise as biomarkers for veterinary diagnosis, and candidates for vaccine production, and treatment agents. Additionally, different treatment or engineering methods could be used to boost the capability of extracellular vesicles. Despite the emerging veterinary interest, EV research has been predominantly human-based. Critical knowledge gaps remain regarding isolation protocols, cargo loading mechanisms, in vivo biodistribution, and species-specific functions. Standardized methods for veterinary EV characterization and validation are lacking. Regulatory uncertainties impede veterinary clinical translation. Advances in fundamental EV biology and technology are needed to propel the veterinary field forward. This review introduces EVs from a veterinary perspective by introducing the latest studies, highlighting their potential while analyzing challenges to motivate expanded veterinary investigation and translation.
Collapse
Affiliation(s)
- Yindi Xiong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Peng Lou
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
12
|
Nathani A, Sun L, Khan I, Aare M, Bagde A, Li Y, Singh M. Combined Role of Interleukin-15 Stimulated Natural Killer Cell-Derived Extracellular Vesicles and Carboplatin in Osimertinib-Resistant H1975 Lung Cancer Cells with EGFR Mutations. Pharmaceutics 2024; 16:83. [PMID: 38258094 PMCID: PMC10821370 DOI: 10.3390/pharmaceutics16010083] [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: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
In this study, we evaluated IL-15 stimulated natural killer cell-derived EVs (NK-EVs) as therapeutic agents in vitro and in vivo in Osimertinib-resistant lung cancer (H1975R) with EGFR mutations (L858R) in combination with carboplatin (CBP). NK-EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis, and atomic force microscopy imaging revealed vesicles with a spherical form and sizes meeting the criteria of exosomal EVs. Further, Western blot studies demonstrated the presence of regular EV markers along with specific NK markers (perforin and granzyme). EVs were also characterized by proteomic analysis, which demonstrated that EVs had proteins for natural killer cell-mediated cytotoxicity (Granzyme B) and T cell activation (perforin and plastin-2). Gene oncology analysis showed that these differentially expressed proteins are involved in programmed cell death and positive regulation of cell death. Further, isolated NK-EVs were cytotoxic to H1975R cells in vitro in 2D and 3D cell cultures. CBP's IC50 was reduced by approximately in 2D and 3D cell cultures when combined with NK-EVs. The EVs were then combined with CBP and administered by i.p. route to H1975R tumor xenografts, and a significant reduction in tumor volume in vivo was observed. Our findings show for the first time that NK-EVs target the PD-L1/PD-1 immunological checkpoint to induce apoptosis and anti-inflammatory response by downregulation of SOD2, PARP, BCL2, SET, NF-κB, and TGF-ß. The ability to isolate functional NK-EVs on a large scale and use them with platinum-based drugs may lead to new clinical applications. The results of the present study suggest the possibility of the combination of NK-cell-derived EVs and CBP as a viable immunochemotherapeutic strategy for resistant cancers.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32304, USA
| | - Islauddin Khan
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (A.N.); (I.K.); (M.A.); (A.B.)
| |
Collapse
|
13
|
Koksal AR, Ekmen N, Aydin Y, Nunez K, Sandow T, Delk M, Moehlen M, Thevenot P, Cohen A, Dash S. A Single-Step Immunocapture Assay to Quantify HCC Exosomes Using the Highly Sensitive Fluorescence Nanoparticle-Tracking Analysis. J Hepatocell Carcinoma 2023; 10:1935-1954. [PMID: 37936599 PMCID: PMC10627088 DOI: 10.2147/jhc.s423043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/07/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Extracellular vesicles could serve as a non-invasive biomarker for early cancer detection. However, limited methods to quantitate cancer-derived vesicles in the native state remain a significant barrier to clinical translation. Aim This research aims to develop a rapid, one-step immunoaffinity approach to quantify HCC exosomes directly from a small serum volume. Methods HCC-derived exosomes in the serum were captured using fluorescent phycoerythrin (PE)-conjugated antibodies targeted to GPC3 and alpha-fetoprotein (AFP). Total and HCC-specific exosomes were then quantified in culture supernatant or patient-derived serums using fluorescence nanoparticle tracking analysis (F-NTA). The performance of HCC exosome quantification in the serum was compared with the tumor size determined by MRI. Results Initially we tested the detection limits of the F-NTA using synthetic fluorescent and non-fluorescent beads. The assay showed an acceptable sensitivity with a detection range of 104-108 particles/mL. Additionally, the combination of immunocapture followed by size-exclusion column purification allows the isolation of smaller-size EVs and quantification by F-NTA. Our assay demonstrated that HCC cell culture releases a significantly higher quantity of GPC3 or GPC3+AFP positive EVs (100-200 particles/cell) compared to non-HCC culture (10-40 particles/cell) (p<0.01 and p<0.05 respectively). The F-NTA enables absolute counting of HCC-specific exosomes in the clinical samples with preserved biological immunoreactivity. The performance of F-NTA was clinically validated in serum from patients ± cirrhosis and with confirmed HCC. F-NTA quantification data show selective enrichment of AFP and GPC3 positive EVs in HCC serum compared to malignancy-free cirrhosis (AUC values for GPC3, AFP, and GPC3/AFP were found 0.79, 0.71, and 0.72 respectively). The MRI-confirmed patient cohort indicated that there was a positive correlation between total tumor size and GPC3-positive exosome concentration (r:0.78 and p<0.001). Conclusion We developed an immunocapture assay that can be used for simultaneous isolation and quantification of HCC-derived exosomes from a small serum volume with high accuracy.
Collapse
Affiliation(s)
- Ali Riza Koksal
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Nergiz Ekmen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Yucel Aydin
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Kelley Nunez
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Tyler Sandow
- Department of Radiology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Molly Delk
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Martin Moehlen
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Paul Thevenot
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
| | - Ari Cohen
- Department of Gastroenterology and Hepatology, Institute of Translational Research, Ochsner Health, New Orleans, LA, USA
- Multi-Organ Transplant Institute, Ochsner Health, New Orleans, LA, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
- Department of Gastroenterology and Hepatology, Tulane University Health Sciences Center, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
| |
Collapse
|
14
|
Deng M, Wu S, Huang P, Liu Y, Li C, Zheng J. Engineered exosomes-based theranostic strategy for tumor metastasis and recurrence. Asian J Pharm Sci 2023; 18:100870. [PMID: 38161784 PMCID: PMC10755545 DOI: 10.1016/j.ajps.2023.100870] [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: 09/02/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024] Open
Abstract
Metastasis-associated processes are the predominant instigator of fatalities linked to cancer, wherein the pivotal role of circulating tumor cells lies in the resurgence of malignant growth. In recent epochs, exosomes, constituents of the extracellular vesicle cohort, have garnered attention within the field of tumor theranostics owing to their inherent attributes encompassing biocompatibility, modifiability, payload capacity, stability, and therapeutic suitability. Nonetheless, the rudimentary functionalities and limited efficacy of unmodified exosomes curtail their prospective utility. In an effort to surmount these shortcomings, intricate methodologies amalgamating nanotechnology with genetic manipulation, chemotherapy, immunotherapy, and optical intervention present themselves as enhanced avenues to surveil and intercede in tumor metastasis and relapse. This review delves into the manifold techniques currently employed to engineer exosomes, with a specific focus on elucidating the interplay between exosomes and the metastatic cascade, alongside the implementation of tailored exosomes in abating tumor metastasis and recurrence. This review not only advances comprehension of the evolving landscape within this domain but also steers the trajectory of forthcoming investigations.
Collapse
Affiliation(s)
- Min Deng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Shuang Wu
- Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Peizheng Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chong Li
- Medical Research Institute, Southwest University, Chongqing 400716, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| |
Collapse
|
15
|
Di Donato M, Medici N, Migliaccio A, Castoria G, Giovannelli P. Exosomes: Emerging Modulators of Pancreatic Cancer Drug Resistance. Cancers (Basel) 2023; 15:4714. [PMID: 37835408 PMCID: PMC10571735 DOI: 10.3390/cancers15194714] [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: 08/10/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Pancreatic cancer (PaC) is one of the most lethal tumors worldwide, difficult to diagnose, and with inadequate therapeutical chances. The most used therapy is gemcitabine, alone or in combination with nanoparticle albumin-bound paclitaxel (nab-paclitaxel), and the multidrug FOLFIRINOX. Unfortunately, PaC develops resistance early, thus reducing the already poor life expectancy of patients. The mechanisms responsible for drug resistance are not fully elucidated, and exosomes seem to be actively involved in this phenomenon, thanks to their ability to transfer molecules regulating this process from drug-resistant to drug-sensitive PaC cells. These extracellular vesicles are released by both normal and cancer cells and seem to be essential mediators of intercellular communications, especially in cancer, where they are secreted at very high numbers. This review illustrates the role of exosomes in PaC drug resistance. This manuscript first provides an overview of the pharmacological approaches used in PaC and, in the last part, focuses on the mechanisms exploited by the exosomes released by cancer cells to induce drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Pia Giovannelli
- Department of Precision Medicine, University of Campania “L.Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
16
|
Bastos N, Castaldo SA, Adem B, Machado JC, Melo CA, Melo SA. SMC3 epigenetic silencing regulates Rab27a expression and drives pancreatic cancer progression. J Transl Med 2023; 21:578. [PMID: 37641131 PMCID: PMC10463307 DOI: 10.1186/s12967-023-04448-1] [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: 05/10/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is expected to soon surpass colorectal cancer as a leading cause of cancer mortality in both males and females in the US, only lagging behind lung cancer. The lethality of PDAC is driven by late diagnosis and inefficient therapies. The complex biology of PDAC involves various cellular components, including exosomes that carry molecular information between cells. Thus, recipient cells can be reprogrammed, impacting tumorigenesis. Rab27a is a GTPase responsible for the last step of exosomes biogenesis. Hence, dissecting the mechanisms that regulate the expression of Rab27a and that control exosomes biogenesis can provide fundamental insights into the molecular underpinnings regulating PDAC progression. METHODS To assess the mechanism that regulates Rab27a expression in PDAC, we used PDAC cell lines. The biological significance of these findings was validated in PDAC genetically engineered mouse models (GEMMs) and human samples. RESULTS In this work we demonstrate in human PDAC samples and GEMMs that Rab27a expression decreases throughout the development of the disease, and that Rab27a knockout promotes disease progression. What is more, we demonstrate that Rab27a expression is epigenetically regulated in PDAC. Treatment with demethylating agents increases Rab27a expression specifically in human PDAC cell lines. We found that SMC3, a component of the cohesin complex, regulates Rab27a expression in PDAC. SMC3 methylation is present in human PDAC specimens and treatment with demethylating agents increases SMC3 expression in human PDAC cell lines. Most importantly, high levels of SMC3 methylation are associated with a worse prognosis in PDAC. Mechanistically, we identified an enhancer region within the Rab27a gene that recruits SMC3, and modulates Rab27a expression. CONCLUSION Overall, we dissected a mechanism that regulates Rab27a expression during PDAC progression and impacts disease prognosis.
Collapse
Affiliation(s)
- Nuno Bastos
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - Stéphanie A Castaldo
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
| | - Bárbara Adem
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - José C Machado
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
| | - Carlos A Melo
- The Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, UK
| | - Sonia A Melo
- i3S - Instituto de Investigação e Inovação Em Saúde, University of Porto, 4200-135, Porto, Portugal.
- Department of Pathology, Faculty of Medicine, University of Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal.
| |
Collapse
|
17
|
Zhong Q, Nie Q, Wu R, Huang Y. Exosomal miR-18a-5p promotes EMT and metastasis of NPC cells via targeting BTG3 and activating the Wnt/β-catenin signaling pathway. Cell Cycle 2023; 22:1544-1562. [PMID: 37287276 PMCID: PMC10361138 DOI: 10.1080/15384101.2023.2216508] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 02/28/2023] [Accepted: 04/26/2023] [Indexed: 06/09/2023] Open
Abstract
This study investigated the underlying mechanism of miR-18a-5p regulating the proliferation, invasion, and metastasis of nasopharyngeal carcinoma (NPC) cells in vitro and in vivo to indicate the pathogenesis of NPC. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was utilized to determine miR-18a-5p expression level in NPC tissues and cell lines. Besides, 2,5-diphenyl-2 H-tetrazolium bromide (MTT) and colony formation assays were employed to detect the effect of miR-18a-5p expression level on NPC cell proliferation. Wound healing and Transwell assays were utilized to detect the effect of miR-18a-5p on NPC cell invasion and migration. The expression levels of epithelial-mesenchymal transition (EMT)-related proteins (Vimentin, N-cadherin, and E-cadherin) were identified by Western blot assay. After collecting exosomes from CNE-2 cells, it was found that exosomal miR-18a-5p secreted from NPC cells promoted NPC cell proliferation, migration, invasion, and EMT, whereas inhibition of miR-18a-5p expression level led to the opposite results. The dual-luciferase reporter assay showed that BTG anti-proliferation factor 3 (BTG3) was the target gene of miR-18a-5p, and BTG3 could overturn the effect of miR-18a-5p on NPC cells. Xenograft mouse model of NPC nude mice showed that miR-18a-5p promoted NPC growth and metastasis in vivo. This study revealed that exosomal miR-18a-5p derived from NPC cells promoted angiogenesis via targeting BTG3 and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Qiong Zhong
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Qihong Nie
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Renrui Wu
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| | - Yun Huang
- Department of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi Province, China
| |
Collapse
|
18
|
Zhang Y, Dou Y, Liu Y, Di M, Bian H, Sun X, Yang Q. Advances in Therapeutic Applications of Extracellular Vesicles. Int J Nanomedicine 2023; 18:3285-3307. [PMID: 37346366 PMCID: PMC10281276 DOI: 10.2147/ijn.s409588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoscale bilayer phospholipid membrane vesicles released by cells. Contained large molecules such as nucleic acid, protein, and lipid, EVs are an integral part of cell communication. The contents of EVs vary based on the cell source and play an important role in both pathological and physiological conditions. EVs can be used as drugs or targets in disease treatment, and changes in the contents of EVs can indicate the progression of diseases. In recent years, with the continuous exploration of the structure, characteristics, and functions of EVs, the potential of engineered EVs for drug delivery and therapy being constantly explored. This review provides a brief overview of the structure, characteristics and functions of EVs, summarizes the advanced application of EVs and outlook on the prospect of it. It is our hope that this review will increase understanding of the current development of medical applications of EVs and help us overcome future challenges.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Mingyuan Di
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Hanming Bian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
19
|
Bhatia R, Chang J, Munoz JL, Walker ND. Forging New Therapeutic Targets: Efforts of Tumor Derived Exosomes to Prepare the Pre-Metastatic Niche for Cancer Cell Dissemination and Dormancy. Biomedicines 2023; 11:1614. [PMID: 37371709 PMCID: PMC10295689 DOI: 10.3390/biomedicines11061614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor-derived exosomes play a multifaceted role in preparing the pre-metastatic niche, promoting cancer dissemination, and regulating cancer cell dormancy. A brief review of three types of cells implicated in metastasis and an overview of other types of extracellular vesicles related to metastasis are described. A central focus of this review is on how exosomes influence cancer progression throughout metastatic disease. Exosomes are crucial mediators of intercellular communication by transferring their cargo to recipient cells, modulating their behavior, and promoting tumor pro-gression. First, their functional role in cancer cell dissemination in the peripheral blood by facilitating the establishment of a pro-angiogenic and pro-inflammatory niche is described during organotro-pism and in lymphatic-mediated metastasis. Second, tumor-derived exosomes can transfer molecular signals that induce cell cycle arrest, dormancy, and survival pathways in disseminated cells, promoting a dormant state are reviewed. Third, several studies highlight exosome involvement in maintaining cellular dormancy in the bone marrow endosteum. Finally, the clinical implications of exosomes as biomarkers or diagnostic tools for cancer progression are also outlined. Understanding the complex interplay between tumor-derived exosomes and the pre-metastatic niche is crucial for developing novel therapeutic strategies to target metastasis and prevent cancer recurrence. To that end, several examples of how exosomes or other nanocarriers are used as a drug delivery system to inhibit cancer metastasis are discussed. Strategies are discussed to alter exosome cargo content for better loading capacity or direct cell targeting by integrins. Further, pre-clinical models or Phase I clinical trials implementing exosomes or other nanocarriers to attack metastatic cancer cells are highlighted.
Collapse
Affiliation(s)
- Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Chang
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| | - Jessian L Munoz
- Division of Perinatal Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nykia D Walker
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| |
Collapse
|
20
|
Wang L, Wang D, Ye Z, Xu J. Engineering Extracellular Vesicles as Delivery Systems in Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300552. [PMID: 37080941 PMCID: PMC10265081 DOI: 10.1002/advs.202300552] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Extracellular vesicles (EVs) are transport vesicles secreted by living cells and released into the extracellular environment. Recent studies have shown that EVs serve as "messengers" in intercellular and inter-organismal communication, in both normal and pathological processes. EVs, as natural nanocarriers, can deliver bioactivators in therapy with their endogenous transport properties. This review article describes the engineering EVs of sources, isolation method, cargo loading, boosting approach, and adjustable targeting of EVs. Furthermore, the review summarizes the recent progress made in EV-based delivery systems applications, including cancer, cardiovascular diseases, liver, kidney, nervous system diseases, and COVID-19 and emphasizes the obstacles and challenges of EV-based therapies and possible strategies.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Di Wang
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Zhaoming Ye
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Jianbin Xu
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| |
Collapse
|
21
|
Peng Z, Tong Z, Ren Z, Ye M, Hu K. Cancer-associated fibroblasts and its derived exosomes: a new perspective for reshaping the tumor microenvironment. Mol Med 2023; 29:66. [PMID: 37217855 DOI: 10.1186/s10020-023-00665-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cells within the tumor microenvironment (TME). They extensively communicate with the other cells. Exosome-packed bioactive molecules derived from CAFs can reshape the TME by interacting with other cells and the extracellular matrix, which adds a new perspective for their clinical application in tumor targeted therapy. An in-depth understanding of the biological characteristics of CAF-derived exosomes (CDEs) is critical for depicting the detailed landscape of the TME and developing tailored therapeutic strategies for cancer treatment. In this review, we have summarized the functional roles of CAFs in the TME, particularly focusing on the extensive communication mediated by CDEs that contain biological molecules such as miRNAs, proteins, metabolites, and other components. In addition, we have also highlighted the prospects for diagnostic and therapeutic applications based on CDEs, which could guide the future development of exosome-targeted anti-tumor drugs.
Collapse
Affiliation(s)
- Zhiwei Peng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Zhiwei Tong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Zihao Ren
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China
| | - Manping Ye
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Anhui, Hefei, 230032, China
| | - Kongwang Hu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, 230022, China.
- Department of General Surgery, Fuyang Affiliated Hospital of Anhui Medical University, Anhui, Fuyang, 236000, China.
| |
Collapse
|
22
|
Colja S, Jovčevska I, Šamec N, Romih R, Zottel A. Sonication is a suitable method for loading nanobody into glioblastoma small extracellular vesicles. Heliyon 2023; 9:e15674. [PMID: 37131433 PMCID: PMC10149266 DOI: 10.1016/j.heliyon.2023.e15674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/04/2023] Open
Abstract
Glioblastoma is one of the deadliest cancers, therefore novel efficient therapeutic approaches are urgently required. One of such are nanobodies, prospective nano-sized bio-drugs with advantageous characteristics. Nanobodies can target intracellular proteins, but to increase their efficiency, the delivery system should be applied. Here, we examined small extracellular vesicles as a delivery system for anti-vimentin nanobody Nb79. Nb79 was loaded in small extracellular vesicles either by incubation with glioblastoma cells, by passive loading into isolated small extracellular vesicles or by sonication of isolated small extracellular vesicles. Small extracellular vesicles secreted by glioblastoma cells were isolated by ultracentrifugation on sucrose cushion. The size distribution and average size of sonicated and non-sonicated small extracellular vesicles were determined by nanoparticle tracking analysis method. The loading of Nb79 into small extracellular vesicles by incubation with cells, passive loading or sonication was confirmed by Western blot and electron microscopy. The effect of small extracellular vesicles on cell survival was determined by WST-1 reagent. Loading of small extracellular vesicles by incubation of cells with Nb79 was unsuccessful and resulted in substantial cell death. On the other hand, as confirmed by Western blot and electron microscopy, sonication is a successful method for obtaining Nb79-loaded small extracellular vesicles. Small extracellular vesicles also had an effect on cell viability. Small extracellular vesicles without Nb79 increased survival of U251 and NCH644 cells for 20-25%, while the Nb79-loaded small extracellular vesicles decreased survival of NCH421k by 11%. We demonstrated that sonication is a suitable method to load nanobodies into exosome, and these small extracellular vesicles could in turn reduce cell survival. The method could be translated also to other applications, such as targeted delivery system of other protein-based drugs.
Collapse
Affiliation(s)
- Sara Colja
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Ivana Jovčevska
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Neja Šamec
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Rok Romih
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Alja Zottel
- Medical Centre for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Corresponding author.
| |
Collapse
|
23
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
24
|
Abstract
Cancer is still a serious health problem globally. Conventional therapies have adverse effects, which affect human life quality. Tumor microenvironment (TME), also known as surrounding stroma, has a contributory role in cancer development. Understanding the interaction between TME and cancer progression is a challenge and helps to develop new therapeutic strategies that neutralize the tracks taken by cancer cells to grow, spread, and resist therapy. Therefore, targeting TME components may be effective in improving tumor therapy. Using nanotechnology for drug delivery is of great interest, where it overcomes some obstacles such as solubility and absorption of drugs and delivering them to the appropriate place of action. The main target of nanotechnology for drug delivery is the ability to differentiate between normal and cancer cells. It can be concluded that TME is an important complementary strategy for the development of anticancer drugs. Multitargeted therapy has better efficient potential than individual therapy against cancer.
Collapse
|
25
|
Metastasis prevention: How to catch metastatic seeds. Biochim Biophys Acta Rev Cancer 2023; 1878:188867. [PMID: 36842768 DOI: 10.1016/j.bbcan.2023.188867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/09/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
Despite considerable advances in the evolution of anticancer therapies, metastasis still remains the main cause of cancer mortality. Therefore, current strategies for cancer cure should be redirected towards prevention of metastasis. Targeting metastatic pathways represents a promising therapeutic opportunity aimed at obstructing tumor cell dissemination and metastatic colonization. In this review, we focus on preclinical studies and clinical trials over the last five years that showed high efficacy in suppressing metastasis through targeting lymph node dissemination, tumor cell extravasation, reactive oxygen species, pre-metastatic niche, exosome machinery, and dormancy.
Collapse
|
26
|
Xu X, Xiang Y, Yang Y, Liu K, Cui Z, Tong X, Chen J, Hou F, Luo Z. The application of tumor cell-derived vesicles in oncology therapy. Clin Transl Oncol 2023; 25:364-374. [PMID: 36207510 DOI: 10.1007/s12094-022-02966-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Tumor cell-derived vesicles are released by tumor cells, have a phospholipid bilayer, and are widely distributed in various biological fluids. In recent years, it has been found that tumor cell-derived vesicles contain proteins, metabolites and nucleic acids and can be delivered to recipient cells to perform their physiological functions, such as mediating specific intercellular communication, activating or inhibiting signaling pathways, participating in regulating the modulation of tumor microenvironment and influencing tumor development, which can be used for early detection and diagnosis of cancer. In addition, tumor cell-derived vesicles exhibit multiple properties in tumor therapeutic applications and may serve as a new class of delivery systems. In this review, we elaborate on the application of tumor cell-derived vesicles in oncology therapy.
Collapse
Affiliation(s)
- Ximei Xu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China.
| | - Yin Xiang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Yang Yang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Kai Liu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiwei Cui
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Xiaodong Tong
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Junliang Chen
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Fang Hou
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiqiang Luo
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| |
Collapse
|
27
|
Ye H, Tan L, Tu C, Min L. Exosomes in sarcoma: Prospects for clinical applications. Crit Rev Oncol Hematol 2023; 181:103895. [PMID: 36481305 DOI: 10.1016/j.critrevonc.2022.103895] [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: 09/02/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Sarcoma is a group of rare and heterogeneous mesenchymal tumors, prone to late diagnosis and poor prognosis. Exosomes are cell-derived small extracellular vesicles found in most body fluids and contain nucleic acids, proteins, lipids, and other molecules. Qualitative and quantitative changes of exosomes and the contents are associated with sarcoma progression, exhibiting their potential as biomarkers. Exosomes possess the capacity of evading immune responses, bioactivity for trafficking, tumor tropism, and lesion residence. Thus, exosomes could be engineered as tumor-specific vehicles in drugs and RNA delivery systems. Exosomes might also serve as therapeutic targets in targeted therapy and immunotherapy and be involved in chemotherapy resistance. Here, we provide a comprehensive summary of exosome applications in liquid biopsy-based diagnosis and explore their implications in the delivery system, targeted therapy, and chemotherapy resistance of sarcoma. Moreover, challenges in exosome clinical applications are raised and some future research directions are proposed.
Collapse
Affiliation(s)
- Huali Ye
- West China Hospital, West China School of Medicine, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Linyun Tan
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Chongqi Tu
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China
| | - Li Min
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Guoxue Xiang No. 37, Chengdu 610041, Sichuan, People's Republic of China.
| |
Collapse
|
28
|
Wang S, Song X, Wang K, Zheng B, Lin Q, Yu M, Xie L, Chen L, Song X. Plasma exosomal miR-320d, miR-4479, and miR-6763-5p as diagnostic biomarkers in epithelial ovarian cancer. Front Oncol 2022; 12:986343. [PMID: 36591520 PMCID: PMC9795228 DOI: 10.3389/fonc.2022.986343] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Background Exosomal miRNA had been proved as the promising biomarkers for multiple cancers including epithelial ovarian cancer (EOC). This study aimed to validate the diagnostic accuracy of exosomal miR-320d, miR-4479, and miR-6763-5p for EOC. Materials and methods Exosomes isolated from the plasma by ultracentrifugation were verified using TEM, qNano and western blot. MiRNAs sequencing was used to screen out the differential exosomal miRNAs and miR-320d, miR-4479, and miR-6763-5p were selected as candidates, which were further verified by RT-qPCR in 168 healthy donors and 161 primary EOC patients. Besides, the diagnostic accuracy of these three exosomal miRNAs were evaluated using the receiver operating characteristic curve (ROC). Results MiRNAs sequencing revealed 95 differential exosomal miRNAs between EOC patients and healthy donors. Subsequently, exosomal miR-320d, miR-4479, and miR-6763-5p were significantly down regulated in EOC patients compared with healthy controls and benign patients. More importantly, these three miRNAs could serve as circulating diagnostics biomarkers for EOC, possessing areas under the curve (AUC) of 0.6549, 0.7781, and 0.6834, respectively. Moreover, these three exosomal miRNAs levels were closely associated with lymph node metastasis, meanwhile exosomal miR-320d and miR-4479 expression was related to tumor stage. Conclusion Exosomal miR-320d, miR-4479, and miR-6763-5p might serve as potential biomarkers for EOC.
Collapse
Affiliation(s)
- Shiwen Wang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xingguo Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kangyu Wang
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Baibing Zheng
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qinghai Lin
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Miao Yu
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Shandong University, Jinan, Shandong, China
| | - Li Xie
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xianrang Song
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China,Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China,*Correspondence: Xianrang Song,
| |
Collapse
|
29
|
Pszczółkowska B, Olejarz W, Filipek M, Tartas A, Kubiak-Tomaszewska G, Żołnierzak A, Życieńska K, Ginter J, Lorenc T, Brzozowska B. Exosome secretion and cellular response of DU145 and PC3 after exposure to alpha radiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:639-650. [PMID: 36098819 PMCID: PMC9630248 DOI: 10.1007/s00411-022-00991-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Exosomes are spherical membrane nanovesicles secreted from cells, and they play an important role in tumor immune response, metastasis, angiogenesis, and survival. Studies investigating exosomes isolated from cells exposed to photon radiation commonly used in conventional radiotherapy demonstrate the influence of this type of radiation on exosome characteristics and secretion. There is currently no research investigating the effects of densely ionizing particles such as protons and alpha radiation on exosomes. Thus we have evaluated the cellular response of human prostate cancer cells exposed to 0, 2, and 6 Gy of alpha radiation emitted from the Am-241 source. Irradiated PC3 and DU145 cell lines, characterized by differences in radiosensitivity, were studied using apoptosis, LDH, and IL-6 assays. Additionally, the corresponding concentration and size of isolated exosomes were measured using NTA. We found that exposure to ionizing radiation resulted in gross changes in viability and cell damage. There were increased amounts of apoptotic or necrotic cells as a function of radiation dose. We demonstrated that irradiated PC3 cells secrete higher quantities of exosomes compared to DU145 cells. Additionally, we also found no statistical difference in exosome size for control and irradiated cells.
Collapse
Affiliation(s)
- Beata Pszczółkowska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Mateusz Filipek
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Adrianna Tartas
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Grażyna Kubiak-Tomaszewska
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Aleksandra Żołnierzak
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
- Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 1 Banacha Street, Warsaw, 02-097 Poland
| | - Katarzyna Życieńska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Józef Ginter
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| | - Tomasz Lorenc
- 1st Department of Clinical Radiology, Medical University of Warsaw, 5 Chałubińskiego Street, Warsaw, 02-004 Poland
| | - Beata Brzozowska
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, 5 Pasteura Street, Warsaw, 02-093 Poland
| |
Collapse
|
30
|
Jiang Z, Wang H, Mou Y, Li L, Jin W. Functions and clinical applications of exosomes in pancreatic cancer. Mol Biol Rep 2022; 49:11037-11048. [PMID: 36097109 PMCID: PMC9618535 DOI: 10.1007/s11033-022-07765-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/05/2023]
Abstract
Pancreatic cancer (PC) is one of the most malignant tumors and has an abysmal prognosis, with a 5-year survival rate of only 11%. At present, the main clinical dilemmas in PC are the lack of biomarkers and the unsatisfactory therapeutic effects. The treatments for and outcomes of PC have improved, but remain unsatisfactory. Exosomes are nanosized extracellular vesicles, and an increasing number of studies have found that exosomes play an essential role in tumor pathology. In this review, we describe the process of exosome biogenesis, as well as exosome extraction methods and identification strategies, and we then explain in detail the roles and mechanisms of exosomes in invasion, metastasis, chemoresistance and immunosuppression in PC. Finally, we summarize the clinical applications of exosomes. Our observations indicate that exosomes represent a novel direction in the clinical treatment of PC.
Collapse
Affiliation(s)
- Zhichen Jiang
- Department of General Surgery,Devision of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, Zhejiang, China
| | - Huiju Wang
- Department of General Surgery,Devision of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, 310014, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Yiping Mou
- Department of General Surgery,Devision of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, 310014, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Li Li
- Department of General Surgery,Devision of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, 310014, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China
| | - Weiwei Jin
- Department of General Surgery,Devision of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China.
- Key Laboratory of Gastroenterology of Zhejiang Province, 310014, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, Zhejiang, China.
| |
Collapse
|
31
|
Jahangiri B, Saei AK, Obi PO, Asghari N, Lorzadeh S, Hekmatirad S, Rahmati M, Velayatipour F, Asghari MH, Saleem A, Moosavi MA. Exosomes, autophagy and ER stress pathways in human diseases: Cross-regulation and therapeutic approaches. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166484. [PMID: 35811032 DOI: 10.1016/j.bbadis.2022.166484] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/01/2022] [Accepted: 07/03/2022] [Indexed: 02/08/2023]
Abstract
Exosomal release pathway and autophagy together maintain homeostasis and survival of cells under stressful conditions. Autophagy is a catabolic process through which cell entities, such as malformed biomacromolecules and damaged organelles, are degraded and recycled via the lysosomal-dependent pathway. Exosomes, a sub-type of extracellular vesicles (EVs) formed by the inward budding of multivesicular bodies (MVBs), are mostly involved in mediating communication between cells. The unfolded protein response (UPR) is an adaptive response that is activated to sustain survival in the cells faced with the endoplasmic reticulum (ER) stress through a complex network that involves protein synthesis, exosomes secretion and autophagy. Disruption of the critical crosstalk between EVs, UPR and autophagy may be implicated in various human diseases, including cancers and neurodegenerative diseases, yet the molecular mechanism(s) behind the coordination of these communication pathways remains obscure. Here, we review the available information on the mechanisms that control autophagy, ER stress and EV pathways, with the view that a better understanding of their crosstalk and balance may improve our knowledge on the pathogenesis and treatment of human diseases, where these pathways are dysregulated.
Collapse
Affiliation(s)
- Babak Jahangiri
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Ali Kian Saei
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Patience O Obi
- Applied Health Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada; Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg R3T 2N2, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada
| | - Narjes Asghari
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Shirin Hekmatirad
- Department of Pharmacology and Toxicology, School of Medicine, Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Velayatipour
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran
| | - Mohammad Hosseni Asghari
- Department of Pharmacology and Toxicology, School of Medicine, Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ayesha Saleem
- Applied Health Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada; Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg R3T 2N2, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, P.O Box 14965/161, Iran.
| |
Collapse
|
32
|
Sahraei N, Mazloum-Ardakani M, Khoshroo A, Hoseynidokht F, Mohiti J, Moradi A. Electrochemical system designed on a paper platform as a label-free immunosensor for cancer derived exosomes based on a mesoporous carbon foam- ternary nanocomposite. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
33
|
Palazzolo S, Canzonieri V, Rizzolio F. The history of small extracellular vesicles and their implication in cancer drug resistance. Front Oncol 2022; 12:948843. [PMID: 36091133 PMCID: PMC9451101 DOI: 10.3389/fonc.2022.948843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/01/2022] Open
Abstract
Small extracellular vesicles (EVs) in the last 20 years are demonstrated to possess promising properties as potential new drug delivery systems, biomarkers, and therapeutic targets. Moreover, EVs are described to be involved in the most important steps of tumor development and progression including drug resistance. The acquired or intrinsic capacity of cancer cells to resist chemotherapies is one of the greatest obstacles to overcome to improve the prognosis of many patients. EVs are involved in this mechanism by exporting the drugs outside the cells and transferring the drug efflux pumps and miRNAs in recipient cells, in turn inducing drug resistance. In this mini-review, the main mechanisms by which EVs are involved in drug resistance are described, giving a rapid and clear overview of the field to the readers.
Collapse
Affiliation(s)
- Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
- Department of Molecular Science and Nanosystems, Ca’ Foscary University, Venice, Italy
| |
Collapse
|
34
|
Nigri J, Leca J, Tubiana SS, Finetti P, Guillaumond F, Martinez S, Lac S, Iovanna JL, Audebert S, Camoin L, Vasseur S, Bertucci F, Tomasini R. CD9 mediates the uptake of extracellular vesicles from cancer-associated fibroblasts that promote pancreatic cancer cell aggressiveness. Sci Signal 2022; 15:eabg8191. [PMID: 35917363 DOI: 10.1126/scisignal.abg8191] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), signaling from stromal cells is implicated in metastatic progression. Tumor-stroma cross-talk is often mediated through extracellular vesicles (EVs). We previously reported that EVs derived from cancer-associated stromal fibroblasts (CAFs) that are abundant in annexin A6 (ANXA6+ EVs) support tumor cell aggressiveness in PDAC. Here, we found that the cell surface glycoprotein and tetraspanin CD9 is a key component of CAF-derived ANXA6+ EVs for mediating this cross-talk. CD9 was abundant on the surface of ANXA6+ CAFs isolated from patient PDAC samples and from various mouse models of PDAC. CD9 colocalized with CAF markers in the tumor stroma, and CD9 abundance correlated with tumor stage. Blocking CD9 impaired the uptake of ANXA6+ EVs into cultured PDAC cells. Signaling pathway arrays and further analyses revealed that the uptake of CD9+ANXA6+ EVs induced mitogen-activated protein kinase (MAPK) pathway activity, cell migration, and epithelial-to-mesenchymal transition (EMT). Blocking either CD9 or p38 MAPK signaling impaired CD9+ANXA6+ EV-induced cell migration and EMT in PDAC cells. Analysis of bioinformatic datasets indicated that CD9 abundance was an independent marker of poor prognosis in patients with PDAC. Our findings suggest that CD9-mediated stromal cell signaling promotes PDAC progression.
Collapse
Affiliation(s)
- Jérémy Nigri
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Julie Leca
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sarah-Simha Tubiana
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Pascal Finetti
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Fabienne Guillaumond
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Sébastien Martinez
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France.,Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Sophie Lac
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Juan L Iovanna
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - Stéphane Audebert
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France.,Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Protéomique, Marseille, France
| | - Luc Camoin
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France.,Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Protéomique, Marseille, France
| | - Sophie Vasseur
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| | - François Bertucci
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France.,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Richard Tomasini
- INSERM, U1068, Cancer Research Center of Marseille, Institut Paoli-Calmettes, CNRS, UMR7258, University Aix-Marseille, Marseille, France
| |
Collapse
|
35
|
Abstract
Exosomes are a type of extracellular vesicles secreted by cells in normal or pathological conditions for cell-cell communication. With immunomodulatory characteristics and potential therapeutic properties, immune-cell-derived exosomes play an important role in cancer therapy. They express various antigens on their surface, which can be employed for antigen presentation, immunological activation, and metabolic regulation, leading to the killing of cancerous cells. In addition, immune-cell-derived exosomes have received extensive attention as a drug delivery platform in effective antitumor therapy due to their excellent biocompatibility, low immunogenicity, and high loading capacity. In this review, the biological and therapeutic characteristics of immune-cell-derived exosomes are comprehensively outlined. The antitumor mechanism of exosomes secreted by immune cells, including macrophages, dendritic cells, T cells, B cells, and natural killer cells, are systematically summarized. Moreover, the applications of immune-cell-derived exosomes as nanocarriers to transport antitumor agents (chemotherapeutic drugs, genes, proteins, etc.) are discussed. More importantly, the existing challenges of immune-cell-derived exosomes are pointed out, and their antitumor potentials are also discussed.
Collapse
Affiliation(s)
- Yongmei Zhao
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales 2145, Australia
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong 226019, China
| |
Collapse
|
36
|
Babaker MA, Aljoud FA, Alkhilaiwi F, Algarni A, Ahmed A, Khan MI, Saadeldin IM, Alzahrani FA. The Therapeutic Potential of Milk Extracellular Vesicles on Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23126812. [PMID: 35743255 PMCID: PMC9224713 DOI: 10.3390/ijms23126812] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer remains one of the leading prevalent cancers in the world and is the fourth most common cause of death from cancer. Unfortunately, the currently utilized chemotherapies fail in selectively targeting cancer cells and cause harm to healthy cells, which results in profound side effects. Researchers are focused on developing anti-cancer targeted medications, which is essential to making them safer, more effective, and more selective and to maximizing their therapeutic benefits. Milk-derived extracellular vesicles (EVs) from camels and cows have attracted much attention as a natural substitute product that effectively suppresses a wide range of tumor cells. This review sheds light on the biogenesis, methods of isolation, characterization, and molecular composition of milk EVs as well as the therapeutic potentials of milk EVs on colorectal cancer.
Collapse
Affiliation(s)
- Manal A. Babaker
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Chemistry, Faculty of Science, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Fadwa A. Aljoud
- Regenerative Medicine Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.); (F.A.)
| | - Faris Alkhilaiwi
- Regenerative Medicine Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.); (F.A.)
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulrahman Algarni
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 73221, Saudi Arabia;
| | - Asif Ahmed
- MirZyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Birmingham B7 4BB, UK;
- School of Health Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Mohammad Imran Khan
- Centre of Artificial Intelligence in Precision Medicines (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Islam M. Saadeldin
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Correspondence: (I.M.S.); (F.A.A.)
| | - Faisal A. Alzahrani
- MirZyme Therapeutics, Innovation Birmingham Campus, Faraday Wharf, Birmingham B7 4BB, UK;
- Centre of Artificial Intelligence in Precision Medicines (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Embryonic Stem Cells Unit, Department of Biochemistry, Faculty of Science, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: (I.M.S.); (F.A.A.)
| |
Collapse
|
37
|
Yang L, Huang S, Zhang Z, Liu Z, Zhang L. Roles and Applications of Red Blood Cell-Derived Extracellular Vesicles in Health and Diseases. Int J Mol Sci 2022; 23:ijms23115927. [PMID: 35682606 PMCID: PMC9180222 DOI: 10.3390/ijms23115927] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/10/2022] Open
Abstract
Red blood cell-derived extracellular vesicles (RBCEVs) are vesicles naturally produced by red blood cells and play multiple roles such as acting as cell-to-cell communication messengers in both normal physiological and diseased states. RBCEVs are highly promising delivery vehicles for therapeutic agents such as biomolecules and nucleic acids as they are easy to source, safe, and versatile. RBCEVs autonomously target the liver and pass the blood-brain barrier into the brain, which is highly valuable for the treatment of liver and brain diseases. RBCEVs can be modified by various functional units, including various functional molecules and nanoparticles, to improve their active targeting capabilities for tumors or other sites. Moreover, the RBCEV level is significantly shifted in many diseased states; hence, they can also serve as important biomarkers for disease diagnoses. It is clear that RBCEVs have considerable potential in multiple medical applications. In this review, we briefly introduce the biological roles of RBCEVs, presented interesting advances in RBCEV applications, and discuss several challenges that need to be addressed for their clinical translation.
Collapse
Affiliation(s)
- Lan Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Shiqi Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Zhenmi Liu
- Med-X Center for Materials, West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Ling Zhang
- Med-X Center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
- Correspondence:
| |
Collapse
|
38
|
Salomon C, Das S, Erdbrügger U, Kalluri R, Kiang Lim S, Olefsky JM, Rice GE, Sahoo S, Andy Tao W, Vader P, Wang Q, Weaver AM. Extracellular Vesicles and Their Emerging Roles as Cellular Messengers in Endocrinology: An Endocrine Society Scientific Statement. Endocr Rev 2022; 43:441-468. [PMID: 35552682 PMCID: PMC10686249 DOI: 10.1210/endrev/bnac009] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 12/15/2022]
Abstract
During the last decade, there has been great interest in elucidating the biological role of extracellular vesicles (EVs), particularly, their hormone-like role in cell-to-cell communication. The field of endocrinology is uniquely placed to provide insight into the functions of EVs, which are secreted from all cells into biological fluids and carry endocrine signals to engage in paracellular and distal interactions. EVs are a heterogeneous population of membrane-bound vesicles of varying size, content, and bioactivity. EVs are specifically packaged with signaling molecules, including lipids, proteins, and nucleic acids, and are released via exocytosis into biofluid compartments. EVs regulate the activity of both proximal and distal target cells, including translational activity, metabolism, growth, and development. As such, EVs signaling represents an integral pathway mediating intercellular communication. Moreover, as the content of EVs is cell-type specific, it is a "fingerprint" of the releasing cell and its metabolic status. Recently, changes in the profile of EV and bioactivity have been described in several endocrine-related conditions including diabetes, obesity, cardiovascular diseases, and cancer. The goal of this statement is to highlight relevant aspects of EV research and their potential role in the field of endocrinology.
Collapse
Affiliation(s)
- Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Saumya Das
- Cardiovascular Research Center of Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Uta Erdbrügger
- Department of Medicine, Nephrology Division, University of Virginia, Charlottesville, VA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Jerrold M Olefsky
- Department of Medicine, University of California-San Diego, La Jolla, CA, USA
| | | | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Pieter Vader
- CDL Research, Division LAB, UMC Utrecht, Utrecht, the Netherlands Faculty of Medicine, Utrecht University, Utrecht, the Netherlands; Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, The Netherlands
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
39
|
Sun H, Bhandari K, Burrola S, Wu J, Ding WQ. Pancreatic Ductal Cell-Derived Extracellular Vesicles Are Effective Drug Carriers to Enhance Paclitaxel's Efficacy in Pancreatic Cancer Cells through Clathrin-Mediated Endocytosis. Int J Mol Sci 2022; 23:4773. [PMID: 35563165 PMCID: PMC9099870 DOI: 10.3390/ijms23094773] [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: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/25/2022] Open
Abstract
Chemo-resistance challenges the clinical management of pancreatic ductal adenocarcinoma (PDAC). A limited admittance of chemotherapeutics to PDAC tissues is a key obstacle in chemotherapy of the malignancy. An enhanced uptake of drugs into PDAC cells is required for a more effective treatment. Extracellular vesicles (EVs), especially small EVs (sEVs), have emerged as drug carriers for delivering chemotherapeutics due to their low immunogenicity and propensity for homing toward tumor cells. The present study evaluated sEVs derived from six different human cell lines as carriers for paclitaxel (PTX). The encapsulation of the chemotherapeutics was achieved using incubation, sonication and electroporation. The cytotoxicity of the EV drugs was evaluated by MTS assay. While sonication led to a higher efficiency of drug loading than incubation and electroporation, PTX loaded through incubation with HPNE-derived sEVs (HI-PTX) was the most efficacious in killing PDAC cells. Furthermore, HI-PTX was taken up by PDAC cells more efficiently than other EV drugs, implying that the efficacy of HI-PTX is associated with its efficient uptake. This was supported by the observation that the cytotoxicity and uptake of HI-PTX is mediated via the clathrin-dependent endocytosis. Our results indicate that the hTERT-HPNE cell-derived EVs are effective drug carriers to enhance paclitaxel's efficacy in PDAC cells.
Collapse
Affiliation(s)
- Haoyao Sun
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China;
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (S.B.)
| | - Kritisha Bhandari
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (S.B.)
| | - Stephanie Burrola
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (S.B.)
| | - Jinchang Wu
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China;
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (S.B.)
| |
Collapse
|
40
|
Chen J, Cao F, Cao Y, Wei S, Zhu X, Xing W. Targeted Therapy of Lung Adenocarcinoma by the Nanoplatform Based on Milk Exosomes Loaded with Paclitaxel. J Biomed Nanotechnol 2022; 18:1075-1083. [PMID: 35854459 DOI: 10.1166/jbn.2022.3278] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lung cancer is the most common cancer throughout the world. Currently, most lung cancer therapies are still limited by serious side effects caused. This paper reports a biocompatible drug delivery system that utilizes milk-derived exosomes to deliver paclitaxel to treat lung adenocarcinoma. First, milk-derived exosomes were modified with integrin αVβ₃, αVβ5-binding peptide iRGD so that they could successfully target lung adenocarcinoma cells. Then, iRGD modified exosomes were loaded with paclitaxel (PAC) via electroporation and used for tumor therapy. These modified exosomes proved effective in killing lung adenocarcinoma cells, and the exosome-based nanoplatform showed no obvious toxicity to normal cells. Further more, the exosome-based nanoplatform could effectively penetrate the interior of the 3D tumor sphere, reaching more tumor cells and demonstrating that it is a promising tool for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Junge Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Fengqiang Cao
- Graduate School of Bio-Applications Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan
| | - Yang Cao
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Shujin Wei
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Xiurui Zhu
- Department of Cardiothoracic Surgery, School of Medicine, Stanford University, California, 94305, USA
| | - Wanli Xing
- School of Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
41
|
Liu K, Gao X, Kang B, Liu Y, Wang D, Wang Y. The Role of Tumor Stem Cell Exosomes in Cancer Invasion and Metastasis. Front Oncol 2022; 12:836548. [PMID: 35350566 PMCID: PMC8958025 DOI: 10.3389/fonc.2022.836548] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Exosomes are lipid membrane bilayer-encapsulated vesicles secreted by cells into the extracellular space. They carry abundant inclusions (such as nucleic acids, proteins, and lipids) that play pivotal roles in intercellular communication. Tumor stem cells are capable of self-renewal and are crucial for survival, proliferation, drug resistance, metastasis, and recurrence of tumors. The miRNAs (microRNAs) in exosomes have various functions, such as participating in inflammatory response, cell migration, proliferation, apoptosis, autophagy, and epithelial-mesenchymal transition. Tumor stem cells secrete exosomes that act as important messengers involved in various tumor processes and several studies provide increasing evidence supporting the importance of these exosomes in tumor recurrence and metastasis. This review primarily focuses on the production and secretion of exosomes from tumors and tumor stem cells and their effects on cancer progression. Cancer stem cancer derived exosome play an important massager in the tumor microenvironment. It also emphasizes on the study of tumor stem cell exosomes in the light of cancer metastasis and recurrence aiming to provide valuable insights and novel perspectives, which could be beneficial for developing effective diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Kun Liu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Xin Gao
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Baoqiang Kang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| | - Yunpeng Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Dingding Wang
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, ChangChun, China
| |
Collapse
|
42
|
Shi Z, Huang K, Li Z, Niu Y, Jiang L. Evaluating the expression of tumorigenic long noncoding RNAs in circulating exosomes isolated from non-small-cell lung cancer patients. Biomark Med 2022; 16:241-251. [PMID: 35209738 DOI: 10.2217/bmm-2021-0930] [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: 11/21/2022] Open
Abstract
Aim: To evaluate the correlation of long noncoding RNAs (lncRNAs) expression in circulating exosomes and the cancerous and noncancerous tissues in patients with non-small-cell lung carcinoma. Methods: The relative expression of the four lncRNAs including LUADT1, MALAT1, NEAT1 and MIAT between tumor tissue, adjacent noncancerous tissues and circulating exosomes were evaluated by quantitative reverse transcription PCR. Results & conclusion: The relative expression of the lncRNAs, including LUADT1, MALAT1 and NEAT1, was upregulated and MIAT was downregulated in tumor tissue compared with noncancerous tissue samples. The expression of lncRNAs in circulating exosomes was not significantly different from cancerous tissue. Our results indicate that the studied exosomal lncRNAs have a good potential to be further evaluated as prognostic/diagnostic biomarkers in patients with non-small-cell lung cancer.
Collapse
Affiliation(s)
- Zhenshan Shi
- Department of Oncology, People's Hospital of Bozhou, Bozhou, Anhui, 236800, PR China
| | - Kaicheng Huang
- Department of Geriatrics, Haikou People's Hospital, Haikou, Hainan, 570208, PR China
| | - Zheng Li
- Department of Second Thoracic Surgery, Anhui Chest Hospital, Hefei, Anhui, 230000, PR China
| | - Yanli Niu
- Department of Operation Room, Xiangyang No 1 People's Hospital, Affiliated Hospital of Hubei University of Medicne, Xiangyang, Hube, 441000, PR China
| | - Lihao Jiang
- Department of Oncology, The People's Hospital of Dazu, Chongqing, Chongqing, 402360, PR China
| |
Collapse
|
43
|
Rizk NI, Abulsoud AI, Kamal MM, Kassem DH, Hamdy NM. Exosomal-long non-coding RNAs journey in colorectal cancer: Evil and goodness faces of key players. Life Sci 2022; 292:120325. [PMID: 35031258 DOI: 10.1016/j.lfs.2022.120325] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/07/2023]
Abstract
Exosomes are nano-vesicles (NVs) secreted by cells and take part in cell-cell communications. Lately, these exosomes were proved to have dual faces in cancer. Actually, they can contribute to carcinogenesis through epithelial-mesenchymal transition (EMT), angiogenesis, metastasis and tumor microenvironment (TME) of various cancers, including colorectal cancer (CRC). On the other hand, they can be potential targets for cancer treatment. CRC is one of the most frequent tumors worldwide, with incidence rates rising in the recent decades. In its early stage, CRC is asymptomatic with poor treatment outcomes. Therefore, finding a non-invasive, early diagnostic biomarker tool and/or suitable defender to combat CRC is mandatory. Exosomes provide enrichment and safe setting for their cargos non-coding RNAs (ncRNAs) and proteins, whose expression levels can be upregulated ordown-regulated in cancer. Hence, exosomes can be used as diagnostic and/or prognostic tools for cancer. Moreover, exosomes can provide a novel potential therapeutic modality for tumors via loading with specific chemotherapeutic agents, with the advantage of possible tumor targeting. In this review, we will try to collect and address recent studies concerned with exosomes and their cargos' implications for CRC diagnosis and/or hopefully, treatment.
Collapse
Affiliation(s)
- Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy (Boys Branch), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohamed M Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt; The Centre for Drug Research and Development, Faculty of Pharmacy, BUE, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
44
|
Ruivo CF, Bastos N, Adem B, Batista I, Duraes C, Melo CA, Castaldo SA, Campos‐Laborie F, Moutinho-Ribeiro P, Morão B, Costa-Pinto A, Silva S, Osorio H, Ciordia S, Costa JL, Goodrich D, Cavadas B, Pereira L, Kouzarides T, Macedo G, Maio R, Carneiro F, Cravo M, Kalluri R, Machado JC, Melo SA. Extracellular Vesicles from Pancreatic Cancer Stem Cells Lead an Intratumor Communication Network (EVNet) to fuel tumour progression. Gut 2022; 71:gutjnl-2021-324994. [PMID: 35012996 PMCID: PMC9271144 DOI: 10.1136/gutjnl-2021-324994] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Intratumor heterogeneity drives cancer progression and therapy resistance. However, it has yet to be determined whether and how subpopulations of cancer cells interact and how this interaction affects the tumour. DESIGN We have studied the spontaneous flow of extracellular vesicles (EVs) between subpopulations of cancer cells: cancer stem cells (CSC) and non-stem cancer cells (NSCC). To determine the biological significance of the most frequent communication route, we used pancreatic ductal adenocarcinoma (PDAC) orthotopic models, patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMMs). RESULTS We demonstrate that PDAC tumours establish an organised communication network between subpopulations of cancer cells using EVs called the EVNet). The EVNet is plastic and reshapes in response to its environment. Communication within the EVNet occurs preferentially from CSC to NSCC. Inhibition of this communication route by impairing Rab27a function in orthotopic xenographs, GEMMs and PDXs is sufficient to hamper tumour growth and phenocopies the inhibition of communication in the whole tumour. Mechanistically, we provide evidence that CSC EVs use agrin protein to promote Yes1 associated transcriptional regulator (YAP) activation via LDL receptor related protein 4 (LRP-4). Ex vivo treatment of PDXs with antiagrin significantly impairs proliferation and decreases the levels of activated YAP.Patients with high levels of agrin and low inactive YAP show worse disease-free survival. In addition, patients with a higher number of circulating agrin+ EVs show a significant increased risk of disease progression. CONCLUSION PDAC tumours establish a cooperation network mediated by EVs that is led by CSC and agrin, which allows tumours to adapt and thrive. Targeting agrin could make targeted therapy possible for patients with PDAC and has a significant impact on CSC that feeds the tumour and is at the centre of therapy resistance.
Collapse
Affiliation(s)
- Carolina F Ruivo
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- ICBAS Instituto de Ciencias Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Nuno Bastos
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- ICBAS Instituto de Ciencias Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Barbara Adem
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- ICBAS Instituto de Ciencias Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ines Batista
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- ICBAS Instituto de Ciencias Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Cecilia Duraes
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Stephanie A Castaldo
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | | | - Pedro Moutinho-Ribeiro
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
- CHUSJ Centro Hospitalar Universitário de São João, Porto, Portugal
| | | | - Ana Costa-Pinto
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Soraia Silva
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Hugo Osorio
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
| | - Sergio Ciordia
- Proteomics Facility, Spanish National Center for Biotechnology, Madrid, Spain
| | - Jose Luis Costa
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
| | | | - Bruno Cavadas
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Luisa Pereira
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Guilherme Macedo
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
- CHUSJ Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Rui Maio
- Hospital Beatriz Ângelo, Loures, Portugal
- Hospital da Luz, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| | - Fatima Carneiro
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
- CHUSJ Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Marília Cravo
- Hospital da Luz, Lisbon, Portugal
- FMUL Faculty of Medicine University of Lisbon, Lisbon, Portugal
| | - Raghu Kalluri
- Cancer Biology, University Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jose Carlos Machado
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
| | - Sonia A Melo
- i3S Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- IPATIMUP Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- FMUP Faculty of Medicine University of Porto, Porto, Portugal
| |
Collapse
|
45
|
Boyne C, Lennox D, Beech O, Powis SJ, Kumar P. What Is the Role of HLA-I on Cancer Derived Extracellular Vesicles? Defining the Challenges in Characterisation and Potential Uses of This Ligandome. Int J Mol Sci 2021; 22:ijms222413554. [PMID: 34948350 PMCID: PMC8703738 DOI: 10.3390/ijms222413554] [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: 12/02/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
The Human Leukocyte Antigen class I (HLA-I) system is an essential part of the immune system that is fundamental to the successful activation of cytotoxic lymphocytes, and an effective subsequent immune attack against both pathogen-infected and cancer cells. The importance of cytotoxic T cell activity and ability to detect foreign cancer-related antigenic peptides has recently been highlighted by the successful application of monoclonal antibody-based checkpoint inhibitors as novel immune therapies. Thus, there is an increased interest in fully characterising the repertoire of peptides that are being presented to cytotoxic CD8+ T cells by cancer cells. However, HLA-I is also known to be present on the surface of extracellular vesicles, which are released by most if not all cancer cells. Whilst the peptide ligandome presented by cell surface HLA class I molecules on cancer cells has been studied extensively, the ligandome of extracellular vesicles remains relatively poorly defined. Here, we will describe the current understanding of the HLA-I peptide ligandome and its role on cancer-derived extracellular vesicles, and evaluate the aspects of the system that have the potential to advance immune-based therapeutic approaches for the effective treatment of cancer.
Collapse
|
46
|
Mammes A, Pasquier J, Mammes O, Conti M, Douard R, Loric S. Extracellular vesicles: General features and usefulness in diagnosis and therapeutic management of colorectal cancer. World J Gastrointest Oncol 2021; 13:1561-1598. [PMID: 34853637 PMCID: PMC8603448 DOI: 10.4251/wjgo.v13.i11.1561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/29/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
In the world, among all type of cancers, colorectal cancer (CRC) is the third most commonly diagnosed in males and the second in females. In most of cases, (RP1) patients’ prognosis limitation with malignant tumors can be attributed to delayed diagnosis of the disease. Identification of patients with early-stage disease leads to more effective therapeutic interventions. Therefore, new screening methods and further innovative treatment approaches are mandatory as they may lead to an increase in progression-free and overall survival rates. For the last decade, the interest in extracellular vesicles (EVs) research has exponentially increased as EVs generation appears to be a universal feature of every cell that is strongly involved in many mechanisms of cell-cell communication either in physiological or pathological situations. EVs can cargo biomolecules, such as lipids, proteins, nucleic acids and generate transmission signal through the intercellular transfer of their content. By this mechanism, tumor cells can recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. This review intends to cover the most recent literature on the role of EVs production in colorectal normal and cancer tissues. Specific attention is paid to the use of EVs for early CRC diagnosis, follow-up, and prognosis as EVs have come into the spotlight of research as a high potential source of ‘liquid biopsies’. The use of EVs as new targets or nanovectors as drug delivery systems for CRC therapy is also summarized.
Collapse
Affiliation(s)
- Aurelien Mammes
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| | - Jennifer Pasquier
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| | | | - Marc Conti
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
- Metabolism Research Unit, Integracell SAS, Longjumeau 91160, France
| | - Richard Douard
- UCBM, Necker University Hospital, Paris 75015, France
- Gastrointestinal Surgery Department, Clinique Bizet, Paris 75016, France
| | - Sylvain Loric
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| |
Collapse
|
47
|
The Role of Exosomes and Their Applications in Cancer. Int J Mol Sci 2021; 22:ijms222212204. [PMID: 34830085 PMCID: PMC8622108 DOI: 10.3390/ijms222212204] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
Exosomes are very small extracellular vesicles secreted by multiple cell types and are extensively distributed in various biological fluids. Recent research indicated that exosomes can participate in regulating the tumor microenvironment and impacting tumor proliferation and progression. Due to the extensive enrollment in cancer development, exosomes have become a focus of the search for a new therapeutic method for cancer. Exosomes can be utilized for the therapeutic delivery of small molecules, proteins and RNAs to target cancer cells with a high efficiency. Exosome-carried proteins, lipids and nucleic acids are being tested as promising biomarkers for cancer diagnosis and prognosis, even as potential treatment targets for cancer. Moreover, different sources of exosomes exhibit multiple performances in cancer applications. In this review, we elaborate on the specific mechanism by which exosomes affect the communication between tumors and the microenvironment and state the therapeutic and diagnostic applications of exosomes in cancers.
Collapse
|
48
|
High-quality milk exosomes as oral drug delivery system. Biomaterials 2021; 277:121126. [PMID: 34544033 DOI: 10.1016/j.biomaterials.2021.121126] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
Many drugs must be administered intravenously instead of oral administration due to their poor oral bioavailability. The cost of repeated infusion treatment for 6 weeks every year is as high as tens of billions of dollars worldwide. Exosomes are nano-sized (30-150 nm) extracellular vesicles secreted by mammalian cells due to environmental stimulation or self-activation. Milk contains abundant exosomes originated from multiple cellular sources. It has been proved that milk exosomes (MEs) could survive with the strongly acidic conditions in the stomach and degradative conditions in the gut. Furthermore, they can cross biological barriers to reach targeted tissues. The ability of MEs to cross the gastrointestinal barrier makes them as a promising drug delivery tool for oral delivery. This review is devoted to the purification of MEs, their biocompatibility and immunogenicity, and prospects for their use as natural drug carriers for oral administration.
Collapse
|
49
|
Pham CV, Midge S, Barua H, Zhang Y, Ngoc-Gia Nguyen T, Barrero RA, Duan A, Yin W, Jiang G, Hou Y, Zhou S, Wang Y, Xie X, Tran PHL, Xiang D, Duan W. Bovine extracellular vesicles contaminate human extracellular vesicles produced in cell culture conditioned medium when 'exosome-depleted serum' is utilised. Arch Biochem Biophys 2021; 708:108963. [PMID: 34126088 DOI: 10.1016/j.abb.2021.108963] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 12/30/2022]
Abstract
Extracellular vesicles (EVs) are important intercellular communication messengers. Half of the published studies in the field are in vitro cell culture based in which bovine serum in various concentrations and forms is used to facilitate the production of extracellular vesicles. 'Exosome depleted serum' is the type of bovine serum most widely used in the production of human EVs. Herein, we demonstrate that, despite the initial caution raised in 2014 about the persistence of bovine EVs, 'exosome depleted serum' was still used in 46% of publications on human or rodent EVs between 2015 and 2019. Using nanoparticle tracking analysis combined with detergent lysis of vesicles as well as bovine CD9 ELISA, we show that there were approximately 5.33 x 107/mL of bovine EVs remaining in the 'exosome depleted serum'. Importantly, the 'exosome depleted serum' was relatively enriched in small EVs by approximately 2.7-fold relative to the large EVs compared to that in the original serum. Specifically, the percentage of small EVs in total vesicles had increased from the original 48% in the serum before ultracentrifugation to 92% in the 'exosome depleted serum'. Furthermore, the pervasive bovine EVs carried over by the 'exosome depleted serum', even when the lowest concentration (0.5%) was used in cell culture, resulted in a significant contamination of human EVs in cell culture conditioned medium. Our findings indicate that the use 'exosome depleted serum' in cell culture-based studies may introduce artefacts into research examining the function of human and rodent EVs, in particular those involving EV miRNA. Thus, we appeal to the researchers in the EV field to seriously reconsider the practice of using 'exosome depleted serum' in the production of human and other mammalian EVs in vitro.
Collapse
Affiliation(s)
- Cuong Viet Pham
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Snehal Midge
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Hridika Barua
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Yumei Zhang
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Tuong Ngoc-Gia Nguyen
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Roberto A Barrero
- eResearch, Division of Research and Innovation, Queensland University of Technology, 2 George Street, Brisbane City, QLD, 4000, Australia
| | - Andrew Duan
- School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University 27 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Wang Yin
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia
| | - Guoqin Jiang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, 215004, PR China
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Shufeng Zhou
- Department of Chemical Engineering & Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, China
| | - Yiming Wang
- Shanghai OneTar Biomedicine, Shanghai, 201203, China
| | - Xiaoqing Xie
- Shanghai OneTar Biomedicine, Shanghai, 201203, China
| | - Phuong H L Tran
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia.
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, 200127, China; Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China.
| | - Wei Duan
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3216, Australia; Shanghai OneTar-Deakin Joint Laboratory of Personalized Precision Medicine, Shanghai, 201203, China.
| |
Collapse
|
50
|
Heparan sulfate analogues regulate tumor-derived exosome formation that attenuates exosome functions in tumor processes. Int J Biol Macromol 2021; 187:481-491. [PMID: 34298051 DOI: 10.1016/j.ijbiomac.2021.07.110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/03/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022]
Abstract
Heparan sulfate (HS) is involved in many biological activities, including the biogenesis and uptake of exosomes, which are related to the occurrence and development of tumors. This study investigated the role of HS analogues (heparin, low molecular weight heparin, and 6-O-desulfated heparin) in modulating exosome secretion, composition and functions. Exosomes derived from B16F10 cells exposed to different HS analogues were isolated and characterized by TEM, western blotting and Nanosight analyses. The number, size and protein cargo of exosomes secreted by HS analogues-induced B16F10 cells were detected. The findings indicated the reduced tumor-derived exosome secretion and protein cargo as reflected by lower levels of CD63, TSG101, heparinase and IL-6 in exosomes derived from heparin-induced B16F10 cells as compared with 6-O-desulfated heparin-induced tumor cells. Further functional assays demonstrated that exosomes from tumor cells exposed to heparin weakened tumor proliferation, migration and invasion most significantly among various exosomes derived from B16F10 cells treated with different HS analogues. Moreover, the sulfate group at 6-O position of heparan sulfate has been proved to play an important role in tumor-derived exosome formation and functions. This study suggested a vital view to develop more specific and efficient HS-based strategies in cancer treatment for targeting tumor-derived exosomes.
Collapse
|