1
|
Neagoe CXR, Ionică M, Neagoe OC, Trifa AP. The Influence of Microbiota on Breast Cancer: A Review. Cancers (Basel) 2024; 16:3468. [PMID: 39456562 PMCID: PMC11506631 DOI: 10.3390/cancers16203468] [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: 09/14/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Breast cancer remains one of the leading causes of death among women worldwide, and recent research highlights its growing connection to alterations in the microbiota. This review delves into the intricate relationship between microbiotas and breast cancer, exploring its presence in healthy breast tissue, its changes during cancer progression, and its considerable impact on both the tumor microenvironment (TME) and the tumor immune microenvironment (TIME). We extensively analyze how the microbiota influences cancer growth, invasion, metastasis, resistance to drugs, and the evasion of the immune system, with a special focus on its effects on the TIME. Furthermore, we investigate distinct microbial profiles associated with the four primary molecular subtypes of breast cancer, examining how the microbiota in tumor tissues compares with that in adjacent normal tissues. Emerging studies suggest that microbiotas could serve as valuable diagnostic and prognostic biomarkers, as well as targets for therapy. This review emphasizes the urgent need for further research to improve strategies for breast cancer prevention, diagnosis, and treatment. By offering a detailed examination of the microbiota's critical role in breast cancer, this review aims to foster the development of novel microbiota-based approaches for managing the disease.
Collapse
Affiliation(s)
- Cara-Xenia-Rafaela Neagoe
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania;
| | - Mihaela Ionică
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Octavian Constantin Neagoe
- Second Clinic of General Surgery and Surgical Oncology, Emergency Clinical Municipal Hospital, 300079 Timișoara, Romania;
- Second Discipline of Surgical Semiology, First Department of Surgery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
- Breast Surgery Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300079 Timișoara, Romania
| | - Adrian Pavel Trifa
- The Discipline of Genetics, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Genetics, Clinical Hospital of Infectious Diseases and Pneumophthisiology “Dr. Victor Babes” Timisoara, 300041 Timisoara, Romania
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
2
|
Almutairy B, Alzahrani MS, Waggas DS, Alsaab HO. Particular exosomal micro-RNAs and gastrointestinal (GI) cancer cells' roles: Current theories. Exp Cell Res 2024; 442:114278. [PMID: 39383930 DOI: 10.1016/j.yexcr.2024.114278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
A diverse range of gastrointestinal tract disorders are called gastrointestinal (GI) malignancies. The transformation of normal cells into precursor cells, precursor cells into premalignant cells, and premalignant cells into cancerous cells is facilitated by the interaction of many modifiable and non-modifiable risk factors. Developing relevant therapy alternatives based on a better knowledge of the illness's aetiology is essential to enhance patient outcomes. The exosome is crucial in regulating intercellular interaction because it may send molecular signals to nearby or distant cells. Exosomes produced from cancer can introduce a variety of chemicals and vast concentrations of microRNA (miRNA) into the tumour microenvironment. These miRNAs significantly impact immunological evasion, metastasis, apoptosis resistance, and cell growth. Exosomal miRNAs, or exosomal miRNAs, are essential for controlling cancer resistance to apoptosis, according to mounting data. Exosomal miRNAs function as an interaction hub between cancerous cells and the milieu around them, regulating gene expression and various signalling pathways. Our research examines the regulatory function of exosomal miRNAs in mediating interactions between cancer cells and the stromal and immunological cells that make up the surrounding milieu.
Collapse
Affiliation(s)
- Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Dania S Waggas
- Pathological Sciences Department, Fakeeh College for Medical Sciences, Jeddah University, Saudi Arabia.
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| |
Collapse
|
3
|
Brachtlova T, Li J, van der Meulen-Muileman IH, Sluiter F, von Meijenfeldt W, Witte I, Massaar S, van den Oever R, de Vrij J, van Beusechem VW. Quantitative Virus-Associated RNA Detection to Monitor Oncolytic Adenovirus Replication. Int J Mol Sci 2024; 25:6551. [PMID: 38928259 PMCID: PMC11203762 DOI: 10.3390/ijms25126551] [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: 05/14/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Oncolytic adenoviruses are in development as immunotherapeutic agents for solid tumors. Their efficacy is in part dependent on their ability to replicate in tumors. It is, however, difficult to obtain evidence for intratumoral oncolytic adenovirus replication if direct access to the tumor is not possible. Detection of systemic adenovirus DNA, which is sometimes used as a proxy, has limited value because it does not distinguish between the product of intratumoral replication and injected virus that did not replicate. Therefore, we investigated if detection of virus-associated RNA (VA RNA) by RT-qPCR on liquid biopsies could be used as an alternative. We found that VA RNA is expressed in adenovirus-infected cells in a replication-dependent manner and is secreted by these cells in association with extracellular vesicles. This allowed VA RNA detection in the peripheral blood of a preclinical in vivo model carrying adenovirus-injected human tumors and on liquid biopsies from a human clinical trial. Our results confirm that VA RNA detection in liquid biopsies can be used for minimally invasive assessment of oncolytic adenovirus replication in solid tumors in vivo.
Collapse
Affiliation(s)
- Tereza Brachtlova
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
- ORCA Therapeutics BV, 5223 DE ‘s-Hertogenbosch, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| | - Jing Li
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ida H. van der Meulen-Muileman
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Femke Sluiter
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
| | - Willem von Meijenfeldt
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
| | - Isabella Witte
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
| | - Sanne Massaar
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
| | | | - Jeroen de Vrij
- Department of Neurosurgery, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- ExoVectory BV, Biopartner 2 Building, 2333 CH Leiden, The Netherlands
| | - Victor W. van Beusechem
- Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.B.); (J.L.)
- ORCA Therapeutics BV, 5223 DE ‘s-Hertogenbosch, The Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunity, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
4
|
Li X, Jing Z, Li X, Liu L, Xiao X, Zhong Y, Ren Z. The role of exosomes in cancer-related programmed cell death. Immunol Rev 2024; 321:169-180. [PMID: 37950410 DOI: 10.1111/imr.13286] [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] [Indexed: 11/12/2023]
Abstract
Cancer arises from the growth and division of uncontrolled erroneous cells. Programmed cell death (PCD), or regulated cell death (RCD), includes natural processes that eliminate damaged or abnormal cells. Dysregulation of PCD is a hallmark of cancer, as cancer cells often evade cell death and continue to proliferate. Exosomes nanoscale extracellular vesicles secreted by different types of cells carrying a variety of molecules, including nucleic acids, proteins, and lipids, to have indispensable role in the communication between cells, and can influence various cellular processes, including PCD. Exosomes have been shown to modulate PCD in cancer cells by transferring pro- or antideath molecules to neighboring cells. Additionally, exosomes can facilitate the spread of PCD to surrounding cancer cells, making them promising in the treatment of various cancers. The exosomes' diagnostic potential in cancer is also an active area of research. Exosomes can be isolated from a wide range of bodily fluids and tissues, such as blood and urine, and can provide a noninvasive way to monitor cancer progression and treatment response. Furthermore, exosomes have also been employed as a delivery system for therapeutic agents. By engineering exosomes to carry drugs or other therapeutic molecules, they can be targeted specifically to cancer cells, reducing toxicity to healthy tissues. Here, we discussed exosomes in the diagnosis and prevention of cancers, tumor immunotherapy, and drug delivery, as well as in different types of PCD.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Xuejie Li
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Lei Liu
- Department of Ophthalmology, Guangdong provincial People's hospital, Guangzhou, China
| | - Xiang Xiao
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Yifan Zhong
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Zihan Ren
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Lv Y, Sun X. Role of miRNA in pathogenesis, diagnosis, and prognosis in hepatocellular carcinoma. Chem Biol Drug Des 2024; 103:e14352. [PMID: 37726253 DOI: 10.1111/cbdd.14352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and is responsible for the second cancer-related death globally. Many treatment regimens have been developed to cure the disease; however, life expectancy is still low. Therefore, there is an urgent need to explore new selective, specific, and robust diagnosis markers for efficient early recognition of the ailment. Along with the diagnosis, the treatment's effectiveness can be determined by prognostic markers, and miRNAs are excellent tools for the diagnosis and prognosis of HCC. In addition, the altered expression profile of a few miRNAs promotes HCC cell migration and invasion, and selective up- or downregulation of these responsible genes may help mitigate the disorder. On one hand, few of the miRNAs have been found to enhance angiogenesis, a crucial step of tumor growth; on the other hand, upregulation of specific miRNAs is reported to suppress angiogenesis and resulting tumor growth of HCC cells. Exosomal miRNAs have significant implications in promoting angiogenesis, increased endothelial cell permeability, tube formation, and metastasis to hepatic and pulmonary tissues. miRNA also attributes to drug resistance toward chemotherapy and the prevention of autophagy also. Identifying novel miRNA and determining their differential expression in HCC tissue may serve as a potential tool for diagnosis, prognosis, and therapy to enhance the life expectancy and quality of life of HCC patients. In the present review, we have summarized the recent advances in HCC-related research.
Collapse
Affiliation(s)
- Yi Lv
- Hepatobiliary and Pancreatic Surgery, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Xiujuan Sun
- Department of Pathology, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| |
Collapse
|
6
|
Sahoo RK, Tripathi SK, Biswal S, Panda M, Mathapati SS, Biswal BK. Transforming native exosomes to engineered drug vehicles: A smart solution to modern cancer theranostics. Biotechnol J 2024; 19:e2300370. [PMID: 38375578 DOI: 10.1002/biot.202300370] [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: 07/27/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024]
Abstract
Exosomes have been the hidden treasure of the cell in terms of cellular interactions, transportation and therapy. The native exosomes (NEx) secreted by the parent cells hold promising aspects in cancer diagnosis and therapy. NEx has low immunogenicity, high biocompatibility, low toxicity and high stability which enables them to be an ideal prognostic biomarker in cancer diagnosis. However, due to heterogeneity, NEx lacks specificity and accuracy to be used as therapeutic drug delivery vehicle in cancer therapy. Transforming these NEx with their innate structure and multiple receptors to engineered exosomes (EEx) can provide better opportunities in the field of cancer theranostics. The surface of the NEx exhibits numeric receptors which can be modified to pave the direction of its therapeutic drug delivery in cancer therapy. Through surface membrane, EEx can be modified with increased drug loading potentiality and higher target specificity to act as a therapeutic nanocarrier for drug delivery. This review provides insights into promising aspects of NEx as a prognostic biomarker and drug delivery tool along with its need for the transformation to EEx in cancer theranostics. We have also highlighted different methods associated with NEx transformations, their nano-bio interaction with recipient cells and major challenges of EEx for clinical application in cancer theranostics.
Collapse
Affiliation(s)
- Rajeev Kumar Sahoo
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Surya Kant Tripathi
- Lineberger Comprehensive Cancer Centre, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Stuti Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Santosh S Mathapati
- Translational Health Science and Technology Institute Faridabad, Faridabad, Haryana, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| |
Collapse
|
7
|
Fang G, Xu D, Zhang T, Qiu L, Gao X, Wang G, Miao Y. Effects of hsa_circ_0074854 on colorectal cancer progression, construction of a circRNA-miRNA-mRNA network, and analysis of immune infiltration. J Cancer Res Clin Oncol 2023; 149:15439-15456. [PMID: 37644235 PMCID: PMC10620273 DOI: 10.1007/s00432-023-05315-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Circular RNAs have been demonstrated to be closely associated with the onset and metastasis of colorectal cancer. However, the roles and clinical diagnostic value of most circRNAs in colorectal cancer remain unclear. METHODS We detected the differential expression of circRNAs in CRC tissues and cells and investigated their relationship in conjunction with clinical pathological features. Additionally, we performed cellular functional experiments in CRC cell lines to explore the functions of circRNAs. To further validate the potential ceRNA network, qPCR was performed to assess the expression of miRNA and mRNA in CRC cells after differential expression of circRNAs knockdown. Furthermore, database analysis was utilized to explore the relationship between the predicted mRNAs and immune infiltration in CRC. RESULTS Our research findings indicate a positive correlation between hsa_circ_0074854 expression and advanced clinical pathological features, as well as an unfavorable prognosis. Knockdown of hsa_circ_0074854 was observed to inhibit proliferation and migration capabilities of colorectal cancer cells, affecting the cell cycle progression, and simultaneously promoting apoptosis. A competing endogenous RNA mechanism may exist among circRNAs, miRNAs, and mRNAs. Furthermore, the expression of target genes displayed correlations with the abundance of certain immune cells. CONCLUSION We propose a novel ceRNA network and evaluate the interplay between target genes and immune cells, providing novel insights for the diagnosis and targeted therapy of CRC.
Collapse
Affiliation(s)
- Guida Fang
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China
| | - Dalai Xu
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang City, Kangda College of Nanjing Medical University, Lianyungang, 222002, Jiangsu, China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China
| | - Lei Qiu
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang City, Kangda College of Nanjing Medical University, Lianyungang, 222002, Jiangsu, China
| | - Xuzhu Gao
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang City, Kangda College of Nanjing Medical University, Lianyungang, 222002, Jiangsu, China
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, 222002, Jiangsu, China
| | - Gang Wang
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China.
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang City, Kangda College of Nanjing Medical University, Lianyungang, 222002, Jiangsu, China.
| | - Yongchang Miao
- Department of Gastrointestinal Surgery, Clinical College of Lianyungang Second People's Hospital, Bengbu Medical College, Lianyungang, 222002, Jiangsu, China.
- Department of Gastrointestinal Surgery, The Second People's Hospital of Lianyungang City, Kangda College of Nanjing Medical University, Lianyungang, 222002, Jiangsu, China.
| |
Collapse
|
8
|
Sun J, Wu L, Wu M, Liu Q, Cao H. Non-coding RNA therapeutics: Towards a new candidate for arsenic-induced liver disease. Chem Biol Interact 2023; 382:110626. [PMID: 37442288 DOI: 10.1016/j.cbi.2023.110626] [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: 03/26/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Arsenic, a metalloid toxicant, has caused serious environmental pollution and is presently a global health issue. Long-term exposure to arsenic causes diverse organ and system dysfunctions, including liver disease. Arsenic-induced liver disease comprises a spectrum of liver pathologies, ranging from hepatocyte damage, steatosis, fibrosis, to hepatocellular carcinoma. Various mechanisms, including an imbalance in redox reactions, mitochondrial dysfunction and epigenetic changes, participate in the pathogenesis of arsenic-induced liver disease. Altered epigenetic processes involved in its initiation and progression. Dysregulated modulations of non-coding RNAs (ncRNAs), including miRNAs, lncRNAs and circRNAs, exert regulating effects on these processes. Here, we have reviewed the underlying pathogenic mechanisms that lead to progressive arsenic-induced liver disease, and we provide a discussion focusing on the effects of ncRNAs on dysfunctions in intercellular communication and on the activation of hepatic stellate cells and malignant transformation of hepatocytes. Further, we have discussed the roles of ncRNAs in intercellular communication via extracellular vesicles and cytokines, and have provided a perspective for the application of ncRNAs as biomarkers in the early diagnosis and evaluation of the pathogenesis of arsenic-induced liver disease. Further investigations of ncRNAs will help us to understand the nature of arsenic-induced liver disease and to identify biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Meng Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| | - Hong Cao
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Sin SQ, Mohan CD, Goh RMWJ, You M, Nayak SC, Chen L, Sethi G, Rangappa KS, Wang L. Hypoxia signaling in hepatocellular carcinoma: Challenges and therapeutic opportunities. Cancer Metastasis Rev 2023; 42:741-764. [PMID: 36547748 DOI: 10.1007/s10555-022-10071-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers with a relatively high cancer-related mortality. The uncontrolled proliferation of HCC consumes a significant amount of oxygen, causing the development of a hypoxic tumor microenvironment (TME). Hypoxia-inducible factors (HIFs), crucial regulators in the TME, activate several cancer hallmarks leading to the hepatocarcinogenesis of HCC and resistance to current therapeutics. As such, HIFs and their signaling pathways have been explored as potential therapeutic targets for the future management of HCC. This review discusses the current understanding of the structure and function of HIFs and their complex relationship with the various cancer hallmarks. To address tumor hypoxia, this review provides an insight into the various potential novel therapeutic agents for managing HCC, such as hypoxia-activated prodrugs, HIF inhibitors, nanomaterials, antisense oligonucleotides, and natural compounds, that target HIFs/hypoxic signaling pathways in HCC. Because of HCC's relatively high incidence and mortality rates in the past decades, greater efforts should be put in place to explore novel therapeutic approaches to improve the outcome for HCC patients.
Collapse
Affiliation(s)
- Shant Qinxiang Sin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | | | - Mingliang You
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Institute, Hangzhou, 31002, China
- Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 31002, China
| | - Siddaiah Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Lu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Cheng J, Zhang K, Qu C, Peng J, Yang L. Non-Coding RNAs Derived from Extracellular Vesicles Promote Pre-Metastatic Niche Formation and Tumor Distant Metastasis. Cancers (Basel) 2023; 15:cancers15072158. [PMID: 37046819 PMCID: PMC10093357 DOI: 10.3390/cancers15072158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Metastasis is a critical stage of tumor progression, a crucial challenge of clinical therapy, and a major cause of tumor patient death. Numerous studies have confirmed that distant tumor metastasis is dependent on the formation of pre-metastatic niche (PMN). Recent studies have shown that extracellular vesicles (EVs) play an important role in PMN formation. The non-coding RNAs (ncRNAs) derived from EVs mediate PMN formation and tumor-distant metastasis by promoting an inflammatory environment, inhibiting anti-tumor immune response, inducing angiogenesis and permeability, and by microenvironmental reprogramming. Given the stability and high abundance of ncRNAs carried by EVs in body fluids, they have great potential for application in tumor diagnosis as well as targeted interventions. This review focuses on the mechanism of ncRNAs derived from EVs promoting tumor PMN formation and distant metastasis to provide a theoretical reference for strategies to control tumor metastasis.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
| | - Kun Zhang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Chunhui Qu
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410078, China
- Department of Pathology, Xiangya Changde Hospital, Changde 415000, China
| | - Lifang Yang
- Department of Oncology, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410078, China
- Cancer Research Institute, School of Basic Medicine Science, Central South University, Changsha 410078, China
| |
Collapse
|
11
|
Yao M, Liang S, Cheng B. Role of exosomes in hepatocellular carcinoma and the regulation of traditional Chinese medicine. Front Pharmacol 2023; 14:1110922. [PMID: 36733504 PMCID: PMC9886889 DOI: 10.3389/fphar.2023.1110922] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) usually occurs on the basis of chronic liver inflammatory diseases and cirrhosis. The liver microenvironment plays a vital role in the tumor initiation and progression. Exosomes, which are nanometer-sized membrane vesicles are secreted by a number of cell types. Exosomes carry multiple proteins, DNAs and various forms of RNA, and are mediators of cell-cell communication and regulate the tumor microenvironment. In the recent decade, many studies have demonstrated that exosomes are involved in the communication between HCC cells and the stromal cells, including endothelial cells, macrophages, hepatic stellate cells and the immune cells, and serve as a regulator in the tumor proliferation and metastasis, immune evasion and immunotherapy. In addition, exosomes can also be used for the diagnosis and treatment HCC. They can potentially serve as specific biomarkers for early diagnosis and drug delivery vehicles of HCC. Chinese herbal medicine, which is widely used in the prevention and treatment of HCC in China, may regulate the release of exosomes and exosomes-mediated intercellular communication. In this review, we summarized the latest progresses on the role of the exosomes in the initiation, progression and treatment of HCC and the potential value of Traditional Chinese medicine in exosomes-mediated biological behaviors of HCC.
Collapse
Affiliation(s)
- Man Yao
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China
| | - Shufang Liang
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University (The Second Military Medical University), Shanghai, China,Faculty of Traditional Chinese Medicine, Naval Medical University (The Second Military Medical University), Shanghai, China,*Correspondence: Binbin Cheng,
| |
Collapse
|
12
|
Chen X, Ren X, E J, Zhou Y, Bian R. Exosome-transmitted circ IFNGR2 Modulates Ovarian Cancer Metastasis via miR-378/ST5 Axis. Mol Cell Biol 2023; 43:22-42. [PMID: 36720469 PMCID: PMC9937009 DOI: 10.1080/10985549.2022.2160605] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/05/2022] [Indexed: 01/28/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs)-derived exosomes have emerged as a key driver of ovarian cancer (OVCA) tumor progression. The mechanisms behind the specific circular RNA (circRNA) activity encapsulated by CAF-generated exosomes (CAF-exo) requires to be elucidated. Herein, this study selected specific circRNA (hsa_circIFNGR2) molecules and aimed to clarify novel function of CAF-derived exosomal circIFNGR2 on growth, and metastasis of OVCA cells. In this study, we clarified that the exosomes of CAFs originating from human ovarian cancer hindered tumor cell proliferation, metastasis and EMT in vitro. Interestingly, CAFs directly transferred exosomes into OVCA cells to enrich intracellular circIFNGR2 levels. Biologically, activation of exosomal circIFNGR2 blocked cell proliferation, metastasis and EMT. Mechanistically, enhanced circIFNGR2 activated the miR-378/ST5 axis and directly inhibited the malignant evolution of tumor cells. Furthermore, rescue experiments evidenced that circIFNGR2 and ST5 were two essential participants in OVCA, concretely manifested in the co-culture of OVCA cells with exosomes that reversed the effects of intracellular circIFNGR2 and ST5 depletion. Finally, we observed that CAF-exo treatment hindered tumor growth and increased the size and number of metastatic nodules in mice. Our study revealed a previously unknown regulatory pathway whereby CAFs-derived exosomes delivered circIFNGR2 and inhibited the malignant progression of OVCA by circIFNGR2/miR-378/ST5 axis.
Collapse
Affiliation(s)
- Xiaoping Chen
- Obstetrics and Gynecology Department, Yancheng First People's Hospital, Yancheng, China
| | - Xinping Ren
- Obstetrics and Gynecology Department, Yancheng First People's Hospital, Yancheng, China
| | - Jiaoting E
- Obstetrics and Gynecology Department, Yancheng First People's Hospital, Yancheng, China
| | - Yaqi Zhou
- Obstetrics and Gynecology Department, Yancheng First People's Hospital, Yancheng, China
| | - Rongfang Bian
- Obstetrics and Gynecology Department, Yancheng First People's Hospital, Yancheng, China
| |
Collapse
|
13
|
Wani AK, Akhtar N, Sharma A, El-Zahaby SA. Fighting Carcinogenesis with Plant Metabolites by Weakening Proliferative Signaling and Disabling Replicative Immortality Networks of Rapidly Dividing and Invading Cancerous Cells. Curr Drug Deliv 2023; 20:371-386. [PMID: 35422214 DOI: 10.2174/1567201819666220414085606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/18/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cancer, an uncontrolled multistage disease causing swift division of cells, is a leading disease with the highest mortality rate. Cellular heterogeneity, evading growth suppressors, resisting cell death, and replicative immortality drive the tumor progression by resisting the therapeutic action of existing anticancer drugs through a series of intrinsic and extrinsic cellular interactions. The innate cellular mechanisms also regulate the replication process as a fence against proliferative signaling, enabling replicative immortality through telomere dysfunction. AREA COVERED The conventional genotoxic drugs have several off-target and collateral side effects associated with them. Thus, the need for the therapies targeting cyclin-dependent kinases or P13K signaling pathway to expose cancer cells to immune destruction, deactivation of invasion and metastasis, and maintaining cellular energetics is imperative. Compounds with anticancer attributes isolated from plants and rich in alkaloids, terpenes, and polyphenols have proven to be less toxic and highly targetspecific, making them biologically significant. This has opened a gateway for the exploration of more novel plant molecules by signifying their role as anticancer agents in synergy and alone, making them more effective than the existing cytotoxic regimens. EXPERT OPINION In this context, the current review presented recent data on cancer cases around the globe, along with discussing the fundamentals of proliferative signaling and replicative immortality of cancer cells. Recent findings were also highlighted, including antiproliferative and antireplicative action of plant-derived compounds, besides explaining the need for improving drug delivery systems.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab (144411), India
| | - Nahid Akhtar
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Punjab (144411), India
| | - Arun Sharma
- Department of Pharmacy, School of Pharmaceutical Sciences, Lovely Professional University, Punjab (144411), India
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
14
|
Ciernikova S, Sevcikova A, Stevurkova V, Mego M. Tumor microbiome - an integral part of the tumor microenvironment. Front Oncol 2022; 12:1063100. [PMID: 36505811 PMCID: PMC9730887 DOI: 10.3389/fonc.2022.1063100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
The tumor microenvironment (TME) plays a significant role in tumor progression and cancer cell survival. Besides malignant cells and non-malignant components, including immune cells, elements of the extracellular matrix, stromal cells, and endothelial cells, the tumor microbiome is considered to be an integral part of the TME. Mounting evidence from preclinical and clinical studies evaluated the presence of tumor type-specific intratumoral bacteria. Differences in microbiome composition between cancerous tissues and benign controls suggest the importance of the microbiome-based approach. Complex host-microbiota crosstalk within the TME affects tumor cell biology via the regulation of oncogenic pathways, immune response modulation, and interaction with microbiota-derived metabolites. Significantly, the involvement of tumor-associated microbiota in cancer drug metabolism highlights the therapeutic implications. This review aims to summarize current knowledge about the emerging role of tumor microbiome in various types of solid malignancies. The clinical utility of tumor microbiome in cancer progression and treatment is also discussed. Moreover, we provide an overview of clinical trials evaluating the role of tumor microbiome in cancer patients. The research focusing on the communication between the gut and tumor microbiomes may bring new opportunities for targeting the microbiome to increase the efficacy of cancer treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia,*Correspondence: Sona Ciernikova,
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viola Stevurkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
15
|
Cui M, Liu Y, Cheng L, Li T, Deng Y, Liu D. Research progress on anti-ovarian cancer mechanism of miRNA regulating tumor microenvironment. Front Immunol 2022; 13:1050917. [DOI: 10.3389/fimmu.2022.1050917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Ovarian cancer is the most deadly malignancy among women, but its complex pathogenesis is unknown. Most patients with ovarian cancer have a poor prognosis due to high recurrence rates and chemotherapy resistance as well as the lack of effective early diagnostic methods. The tumor microenvironment mainly includes extracellular matrix, CAFs, tumor angiogenesis and immune-associated cells. The interaction between tumor cells and TME plays a key role in tumorigenesis, progression, metastasis and treatment, affecting tumor progression. Therefore, it is significant to find new tumor biomarkers and therapeutic targets. MicroRNAs are non-coding RNAs that post-transcriptionally regulate the expression of target genes and affect a variety of biological processes. Studies have shown that miRNAs regulate tumor development by affecting TME. In this review, we summarize the mechanisms by which miRNAs affect ovarian cancer by regulating TME and highlight the key role of miRNAs in TME, which provides new targets and theoretical basis for ovarian cancer treatment.
Collapse
|
16
|
Zhou X, Chen B, Zhang Z, Huang Y, Li J, Wei Q, Cao D, Ai J. Crosstalk between Tumor-Associated Macrophages and MicroRNAs: A Key Role in Tumor Microenvironment. Int J Mol Sci 2022; 23:13258. [PMID: 36362044 PMCID: PMC9653885 DOI: 10.3390/ijms232113258] [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: 09/17/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
As an in-depth understanding of immunotherapy continues to grow, current anticancer therapy research is increasingly focused on the tumor microenvironment (TME). MicroRNAs (miRNAs) play crucial roles in the regulation of genetic information and expression and mediate interactions between tumor cells and components in the TME, such as tumor-associated macrophages (macrophages). Macrophages are abundant in the TME, and their different polarization directions can promote or inhibit tumor growth and progression. By regulating biological behaviors, such as macrophage recruitment, infiltration, and polarization, miRNAs can affect various molecular pathways to regulate tumor progression and treatment response. In this review, we discuss in detail the effects of macrophages on tumors and the multifaceted effects of miRNAs on macrophages. We also discuss the potential clinical applications and prospects of targeted therapy based on miRNAs, novel clinical biomarkers, and drug delivery systems.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dehong Cao
- Department of Urology, Institute of Urology, Sichuan University, Chengdu 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Tian BW, Han CL, Dong ZR, Tan SY, Wang DX, Li T. Role of Exosomes in Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14164036. [PMID: 36011030 PMCID: PMC9406927 DOI: 10.3390/cancers14164036] [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: 06/14/2022] [Revised: 07/21/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma is one of the most lethal malignancies, having a significantly poor prognosis. Immunotherapy, as an emerging tumor treatment option, provides new hope for many cancer patients. However, a large proportion of patients do not benefit from immunotherapy. As a critical cell-to-cell communication mediator in the tumor immune microenvironment, exosomes may play a unique role in hepatocellular carcinoma immune response and thus affect the efficiency of immunotherapy. In this review, we discuss related research on the roles of exosomes in the current immunotherapy resistance mechanism of hepatocellular carcinoma. Furthermore, we also clarify the excellent predictive value of exosomes and the roles they play in improving immunotherapy efficacy for hepatocellular carcinoma patients. We hope that our review can help readers to gain a more comprehensive understanding of exosomes’ roles in hepatocellular carcinoma immunotherapy. Abstract Hepatocellular carcinoma (HCC) is one of the most lethal malignancies, having a significantly poor prognosis and no sufficiently efficient treatments. Immunotherapy, especially immune checkpoint inhibitors (ICIs), has provided new therapeutic approaches for HCC patients. Nevertheless, most patients with HCC do not benefit from immunotherapy. Exosomes are biologically active lipid bilayer nano-sized vesicles ranging in size from 30 to 150 nm and can be secreted by almost any cell. In the HCC tumor microenvironment (TME), numerous cells are involved in tumor progression, and exosomes—derived from tumor cells and immune cells—exhibit unique composition profiles and act as intercellular communicators by transporting various substances. Showing the dual characteristics of tumor promotion and suppression, exosomes exert multiple functions in shaping tumor immune responses in the crosstalk between tumor cells and surrounding immune cells, mediating immunotherapy resistance by affecting the PD-1/PD-L1 axis or the anti-tumor function of immune cells in the TME. Targeting exosomes or the application of exosomes as therapies is involved in many aspects of HCC immunotherapies (e.g., ICIs, tumor vaccines, and adoptive cell therapy) and may substantially enhance their efficacy. In this review, we discuss the impact of exosomes on the HCC TME and comprehensively summarize the role of exosomes in immunotherapy resistance and therapeutic application. We also discuss the potential of exosomes as biomarkers for predicting the efficacy of immunotherapy to help clinicians in identifying HCC patients who are amenable to immunotherapies.
Collapse
Affiliation(s)
- Bao-Wen Tian
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
| | - Cheng-Long Han
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
| | - Dong-Xu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250000, China
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan 250000, China
- Correspondence: ; Tel./Fax: +86-531-8216-6651
| |
Collapse
|
18
|
Zelli V, Compagnoni C, Capelli R, Corrente A, Di Vito Nolfi M, Zazzeroni F, Alesse E, Tessitore A. Role of exosomal microRNAs in cancer therapy and drug resistance mechanisms: focus on hepatocellular carcinoma. Front Oncol 2022; 12:940056. [PMID: 35912267 PMCID: PMC9334682 DOI: 10.3389/fonc.2022.940056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), defined as intercellular messengers that carry their cargos between cells, are involved in several physiological and pathological processes. These small membranous vesicles are released by most cells and contain biological molecules, including nucleic acids, proteins and lipids, which can modulate signaling pathways of nearby or distant recipient cells. Exosomes, one the most characterized classes of EVs, include, among others, microRNAs (miRNAs), small non-coding RNAs able to regulate the expression of several genes at post-transcriptional level. In cancer, exosomal miRNAs have been shown to influence tumor behavior and reshape tumor microenvironment. Furthermore, their possible involvement in drug resistance mechanisms has become evident in recent years. Hepatocellular carcinoma (HCC) is the major type of liver cancer, accounting for 75-85% of all liver tumors. Although the improvement in HCC treatment approaches, low therapeutic efficacy in patients with intermediate-advanced HCC is mainly related to the development of tumor metastases, high risk of recurrence and drug resistance. Exosomes have been shown to be involved in pathogenesis and progression of HCC, as well as in drug resistance, by regulating processes such as cell proliferation, epithelial-mesenchymal transition and immune response. Herein, we summarize the current knowledge about the involvement of exosomal miRNAs in HCC therapy, highlighting their role as modulators of therapeutic response, particularly chemotherapy and immunotherapy, as well as possible therapeutic tools.
Collapse
Affiliation(s)
- Veronica Zelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- Center for Molecular Diagnostics and Advanced Therapies, University of L’Aquila, L’Aquila, Italy
| | - Chiara Compagnoni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberta Capelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandra Corrente
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- Center for Molecular Diagnostics and Advanced Therapies, University of L’Aquila, L’Aquila, Italy
- *Correspondence: Alessandra Tessitore,
| |
Collapse
|
19
|
Exosomal microRNA-25 released from cancer cells targets SIK1 to promote hepatocellular carcinoma tumorigenesis. Dig Liver Dis 2022; 54:954-963. [PMID: 34384713 DOI: 10.1016/j.dld.2021.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is recognized as a leading cause of cancer-associated fatality worldwide. Our study here aimed to probe the mechanism by which exosomes secreted by CSQT-2, an HCC cell line, affected the progression of HCC. METHODS Exosomes were extracted from CSQT-2 cells. Colony formation, Transwell, sphere formation and flow cytometric analyses were applied to assess cell biological activities. Microarray analysis detected the change of microRNA (miRNA) expression after exosome treatment, followed by RT-qPCR validation. Luciferase reporter was applied to detect the binding between SIK1 and miR-25. Xenograft studies in nude mice manifested tumor growth and metastatic ability of miR-25 and SIK1. RESULTS The exosome treatment enhanced cell malignant phenotype in vitro and tumor growth and liver and lung metastases in vivo. The exosomes elevated miR-25 expression in HCC cells. miR-25 targeted SIK1 which was decreased in the exosomes-treated cells. miR-25 inhibitor reduced cell malignant phenotype and attenuated tumorigenesis and metastasis in vivo. SIK1 silencing reversed the effect of miR-25 inhibitor. The exosome treatment potentiated the Wnt/β-catenin pathway in cells, whereas miR-25 inhibitor blunted the pathway activity. CONCLUSION MiR-25 shuttled through CSQT-2-derived exosomes promoted the development of HCC by reducing SIK1 expression and potentiating the Wnt/β-catenin pathway.
Collapse
|
20
|
Peng L, Wang F, Wang Z, Tan J, Huang L, Tian X, Liu G, Zhou L. Cell-cell communication inference and analysis in the tumour microenvironments from single-cell transcriptomics: data resources and computational strategies. Brief Bioinform 2022; 23:6618236. [PMID: 35753695 DOI: 10.1093/bib/bbac234] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Carcinomas are complex ecosystems composed of cancer, stromal and immune cells. Communication between these cells and their microenvironments induces cancer progression and causes therapy resistance. In order to improve the treatment of cancers, it is essential to quantify crosstalk between and within various cell types in a tumour microenvironment. Focusing on the coordinated expression patterns of ligands and cognate receptors, cell-cell communication can be inferred through ligand-receptor interactions (LRIs). In this manuscript, we carry out the following work: (i) introduce pipeline for ligand-receptor-mediated intercellular communication estimation from single-cell transcriptomics and list a few available LRI-related databases and visualization tools; (ii) demonstrate seven classical intercellular communication scoring strategies, highlight four types of representative intercellular communication inference methods, including network-based approaches, machine learning-based approaches, spatial information-based approaches and other approaches; (iii) summarize the evaluation and validation avenues for intercellular communication inference and analyze the advantages and limitations for the above four types of cell-cell communication methods; (iv) comment several major challenges while provide further research directions for intercellular communication analysis in the tumour microenvironments. We anticipate that this work helps to better understand intercellular crosstalk and to further develop powerful cell-cell communication estimation tools for tumor-targeted therapy.
Collapse
Affiliation(s)
- Lihong Peng
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China.,College of Life Sciences and Chemistry, Hunan University of Technology, 412007, Hunan, China
| | - Feixiang Wang
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Zhao Wang
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Jingwei Tan
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Li Huang
- Academy of Arts and Design, Tsinghua University, 10084, Beijing, China.,The Future Laboratory, Tsinghua University, 10084, Beijing, China
| | - Xiongfei Tian
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Guangyi Liu
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Liqian Zhou
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| |
Collapse
|
21
|
Park K, Veena MS, Shin DS. Key Players of the Immunosuppressive Tumor Microenvironment and Emerging Therapeutic Strategies. Front Cell Dev Biol 2022; 10:830208. [PMID: 35345849 PMCID: PMC8957227 DOI: 10.3389/fcell.2022.830208] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic battlefield for both immune cells and tumor cells. The advent of the immune checkpoint inhibitors (ICI) since 2011, such as the anti-cytotoxic T-lymphocyte associated protein (CTLA)-4 and anti-programmed cell death receptor (PD)-(L)1 antibodies, provided powerful weapons in the arsenal of cancer treatments, demonstrating unprecedented durable responses for patients with many types of advanced cancers. However, the response rate is generally low across tumor types and a substantial number of patients develop acquired resistance. These primary or acquired resistance are attributed to various immunosuppressive elements (soluble and cellular factors) and alternative immune checkpoints in the TME. Therefore, a better understanding of the TME is absolutely essential to develop therapeutic strategies to overcome resistance. Numerous clinical studies are underway using ICIs and additional agents that are tailored to the characteristics of the tumor or the TME. Some of the combination treatments are already approved by the Food and Drug Administration (FDA), such as platinum-doublet chemotherapy, tyrosine kinase inhibitor (TKI) -targeting vascular endothelial growth factor (VEGF) combined with anti-PD-(L)1 antibodies or immuno-immuno combinations (anti-CTLA-4 and anti-PD-1). In this review, we will discuss the key immunosuppressive cells, metabolites, cytokines or chemokines, and hypoxic conditions in the TME that contribute to tumor immune escape and the prospect of relevant clinical trials by targeting these elements in combination with ICIs.
Collapse
Affiliation(s)
- Kevin Park
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Mysore S Veena
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Daniel Sanghoon Shin
- Department of Medicine, Division of Hematology/Oncology, Los Angeles, CA, United States.,VA Greater Los Angeles Healthcare System, University of California, Los Angeles (UCLA), Los Angeles, CA, United States.,Molecular Biology Institute, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
22
|
Fan Q, Yu Y, Zhou Y, Zhang S, Wu C. An emerging role of radiation‑induced exosomes in hepatocellular carcinoma progression and radioresistance (Review). Int J Oncol 2022; 60:46. [PMID: 35266016 PMCID: PMC8923655 DOI: 10.3892/ijo.2022.5336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/01/2022] [Indexed: 11/20/2022] Open
Abstract
The incidence rates of hepatocellular carcinoma (HCC) worldwide are increasing, and the role of radiotherapy is currently under discussion. Radioresistance is one of the most important challenges in the therapy of HCC compared with other local advanced, recurrent and metastatic cancers. The mechanisms of radioresistance are complex and remain to be fully understood; however, extracellular vesicles have been investigated in recent studies. Exosomes, which are 40- to 150-nm extracellular vesicles released by cancer cells, contain multiple pathogenic components, including proteins, nucleic acids and lipids, and play critical functions in cancer progression. Emerging data indicate a diagnosis potential for exosomes in HCC, since radiation-derived exosomes promote radioresistance. Radiation-based therapy alters the contents and components of exosomes, suggesting that exosomes and their components may serve as prognostic and predictive biomarkers to monitor radiation response. Therefore, understanding the roles and mechanisms of exosomes in HCC progression and radiation response during HCC therapy may increase our knowledge concerning the roles of exosomes in radioresistance, and may lead to novel approaches for HCC prognosis and treatment. The current review summarizes recent studies on exosome involvement in HCC and the molecular changes in exosome components during HCC progression. It also discusses the functions of exosomes in HCC therapy, and highlights the importance of exosomes in HCC progression and resistance for the development of novel therapies.
Collapse
Affiliation(s)
- Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Yue Yu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Yueling Zhou
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Sheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Chunli Wu
- Department of Radiation Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
23
|
EGCG Prevents the Onset of an Inflammatory and Cancer-Associated Adipocyte-like Phenotype in Adipose-Derived Mesenchymal Stem/Stromal Cells in Response to the Triple-Negative Breast Cancer Secretome. Nutrients 2022; 14:nu14051099. [PMID: 35268073 PMCID: PMC8912398 DOI: 10.3390/nu14051099] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) cells secretome induces a pro-inflammatory microenvironment within the adipose tissue, which hosts both mature adipocytes and adipose-derived mesenchymal stem/stromal cells (ADMSC). The subsequent acquisition of a cancer-associated adipocyte (CAA)-like phenotype is, however, unknown in ADMSC. While epidemiological studies suggest that consuming a polyphenol-rich diet reduces the incidence of some obesity-related cancers, the chemopreventive impact of green tea-derived epigallocatechin-3-gallate (EGCG) against the cues that trigger the CAA phenotype remain undocumented in ADMSC. Methods: Human ADMSC were exposed to human TNBC-derived MDA-MB-231 conditioned media (TNBC cells secretome) supplemented or not with EGCG. Differential gene expression was assessed through RNA-Seq analysis and confirmed by RT-qPCR. Protein expression levels and the activation status of signal transduction pathways mediators were determined by Western blotting. ADMSC chemotaxis was assessed by a real-time cell migration assay. Results: The TNBC cells secretome induced in ADMSC the expression of the CAA cytokines CCL2, CCL5, IL-1β, and IL-6, and of immunomodulators COX2, HIF-1α, VEGFα, and PD-L1. The epithelial-to-mesenchymal biomarker Snail was found to control the CAA phenotype. EGCG inhibited the induction of CAA genes and the activation status of Smad2 and NF-κB. The induced chemotactic response was also inhibited by EGCG. Conclusion: The induction of an inflammatory and CAA-like phenotype in ADMSC can be triggered by the TNBC cells secretome, while still efficiently prevented by diet-derived polyphenols.
Collapse
|
24
|
Jang JW, Kim JM, Kim HS, Kim JS, Han JW, Lee SK, Nam H, Sung PS, Bae SH, Choi JY, Yoon SK. Diagnostic performance of serum exosomal miRNA-720 in hepatocellular carcinoma. JOURNAL OF LIVER CANCER 2022; 22:30-39. [PMID: 37383532 PMCID: PMC10035706 DOI: 10.17998/jlc.2022.02.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/13/2022] [Accepted: 02/24/2022] [Indexed: 06/30/2023]
Abstract
Background/Aim Hepatocellular carcinoma (HCC) is associated with poor prognosis, largely due to late detection. Highly accurate biomarkers are urgently needed to detect early-stage HCC. Our study aims to explore the diagnostic performance of serum exosomal microRNA (miR)-720 in HCC. Methods Exosomal miRNA was measured via quantitative real-time PCR. A correlation analysis of exosomal miR-720 and tumor or clinico-demographic data of patients with HCC was performed. The receiver operating characteristic (ROC) curve was used to assess the diagnostic capacity of serum exosomal miR-720 for HCC, in comparison with α-fetoprotein (AFP) and prothrombin induced by vitamin K absence or antagonist-II (PIVKA-II). Results MiR-720 was chosen as a potential HCC marker via miR microarray based on significant differential expression between tumor and non-tumor samples. Serum exosomal miR-720 was significantly upregulated in patients with HCC (n=114) versus other liver diseases (control, n=30), with a higher area under the ROC curve (AUC, 0.931) than the other markers. Particularly, serum exosomal miR-720 showed superior performance in diagnosing small HCC (<5 cm; AUC, 0.930) compared with AFP (AUC, 0.802) or PIVKA-II (AUC, 0.718). Exosomal miR-720 levels showed marginal correlation with tumor size. The proportion of elevated miR-720 also increased with intrahepatic tumor stage progression. Unlike AFP or PIVKA-II showing a significant correlation with aminotransferase levels, the exosomal miR-720 level was not affected by aminotransferase levels. Conclusions Serum exosomal miR-720 is an excellent biomarker for the diagnosis of HCC, with better performance than AFP or PIVKA-II. Its diagnostic utility is maintained even in small HCC and is unaffected by aminotransferase levels.
Collapse
Affiliation(s)
- Jeong Won Jang
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Ji Min Kim
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Seon Kim
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Seoub Kim
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Won Han
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Soon Kyu Lee
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Heechul Nam
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Pil Soo Sung
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Si Hyun Bae
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Jong Young Choi
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| | - Seung Kew Yoon
- Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
25
|
Xue D, Han J, Liang Z, Jia L, Liu Y, Tuo H, Peng Y. Current Perspectives on the Unique Roles of Exosomes in Drug Resistance of Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:99-112. [PMID: 35211428 PMCID: PMC8863332 DOI: 10.2147/jhc.s351038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/29/2022] [Indexed: 12/14/2022] Open
Abstract
As a common malignant tumor worldwide, the prognosis of hepatocellular carcinoma (HCC) remains unsatisfactory, even though treatment methods have improved. Despite the developments in traditional chemotherapy and emerging targeted immunotherapy, the problem of recurrence and metastasis of HCC and adverse effects on survival and prognosis are still serious. Drug resistance is a daunting challenge that impedes HCC treatment. Exosomes, a class of extracellular vesicles ranging in size from 30 to 100 nm, have been the focus of recent studies. Exosomes can activate various signaling pathways and regulate the tumor microenvironment with their cargo, which includes functional lipids, proteins, and nucleic acids. Thus, they change the phenotype of recipient cells via exosome-mediated communication. Exosomes secreted by tumors or stromal cells can also transfer drug-resistant traits to other tumor cells. However, their effects on drug resistance in HCC are not completely understood. In this review, we summarize and discuss the underlying relationship between exosomes and drug resistance in HCC. In addition, we also show that exosomes may act as candidate biomarkers for predicting and monitoring drug responses and as potential targets or vectors to reverse the drug resistance of HCC.
Collapse
Affiliation(s)
- Dongdong Xue
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Jingzhao Han
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Ze Liang
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Lin Jia
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Yifan Liu
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
- Department of Graduate School, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Hongfang Tuo
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Yanhui Peng
- Department of Hepatobiliary Surgery, Hebei General Hospital, Shijiazhuang, People’s Republic of China
- Correspondence: Yanhui Peng, Department of Hepatobiliary Surgery, Hebei General Hospital, No. 348 Heping West Road, Xinhua District, Shijiazhuang, 050051, Hebei Province, People’s Republic of China, Tel/Fax +86-311-859 8321, Email
| |
Collapse
|
26
|
Yang R, Wang D, Han S, Gu Y, Li Z, Deng L, Yin A, Gao Y, Li X, Yu Y, Wang X. MiR-206 suppresses the deterioration of intrahepatic cholangiocarcinoma and promotes sensitivity to chemotherapy by inhibiting interactions with stromal CAFs. Int J Biol Sci 2022; 18:43-64. [PMID: 34975317 PMCID: PMC8692143 DOI: 10.7150/ijbs.62602] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Intrahepatic cholangiocarcinoma (iCCA) is a highly malignant subtype of cholangiocarcinoma (CCA) with poor prognosis. In iCCA, the interplay between the stroma and tumor cells results in resistance to adjuvant chemotherapy. Increasing evidence indicates that miR-206 participates in tumor progression, but its role in iCCA is still unclear. The aim of this study was to identify dysregulated miR-206 expression in iCCA and to further explore the underlying mechanism. Methods: MiR-206 expression was proven to be downregulated in iCCA tissues by qPCR, and its correlation with clinical characteristics and prognosis was investigated. iCCA-derived cancer-associated fibroblast cells (CAFs) and normal fibroblast cells (NFs) were isolated and identified. MiR-206 was knocked in or down in CAFs and CCA cells, respectively, to explore the role of miR-206, and coculture of these treated CCAs and CAFs was conducted to explore the effects of miR-206 on their mutual promoting effects. Exosomes carrying miR-206 and an orthotopic mouse model were used to determine the inhibitory effects of miR-206 on iCCA deterioration in vivo. Results: We confirmed that miR-206 is a suppressor of iCCA. Overexpressing miR-206 in CCA cells inhibited cell proliferation, migration and invasion. When cocultured with CCA cells, NFs downregulated miR-206 expression, and NFs were susceptible to transforming into CAFs. Moreover, CAFs promoted CCA cell malignant behaviors and gemcitabine resistance. Overexpressing miR-206 in CAFs or CCA cells inhibited this mutual promoting effect. Additionally, when delivered by exosomes, miR-206 suppressed tumor deterioration. And combined with gemcitabine, this treatment resulted in a longer survival time. Conclusion: Our study explained that the interaction between CCA cells and CAFs promoted iCCA deterioration. As a suppressive factor, miR-206 inhibited aggressive characteristics and gemcitabine resistance by interfering with this mutual promoting effect. This research elucidated the molecular mechanism underlying the unfavorable chemotherapeutic response of patients with iCCA, which provided a promising target for iCCA treatment.
Collapse
Affiliation(s)
- Renjie Yang
- School of Medicine, Southeast University, Nanjing, China.,Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Dong Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Shen Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yichao Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Zhi Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Lei Deng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Aihong Yin
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yue Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- School of Medicine, Southeast University, Nanjing, China.,Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| |
Collapse
|
27
|
Li C, Zhou T, Chen J, Li R, Chen H, Luo S, Chen D, Cai C, Li W. The role of Exosomal miRNAs in cancer. J Transl Med 2022; 20:6. [PMID: 34980158 PMCID: PMC8722109 DOI: 10.1186/s12967-021-03215-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/22/2021] [Indexed: 02/08/2023] Open
Abstract
Exosomal miRNAs have attracted much attention due to their critical role in regulating genes and the altered expression of miRNAs in virtually all cancers affecting humans (Sun et al. in Mol Cancer 17(1):14, 2018). Exosomal miRNAs modulate processes that interfere with cancer immunity and microenvironment, and are significantly involved in tumor growth, invasion, metastasis, angiogenesis and drug resistance. Fully investigating the detailed mechanism of miRNAs in the occurrence and development of various cancers could help not only in the treatment of cancers but also in the prevention of malignant diseases. The current review highlighted recently published advances regarding cancer-derived exosomes, e.g., sorting and delivery mechanisms for RNAs. Exosomal miRNAs that modulate cancer cell-to-cell communication, impacting tumor growth, angiogenesis, metastasis and multiple biological features, were discussed. Finally, the potential role of exosomal miRNAs as diagnostic and prognostic molecular markers was summarized, as well as their usefulness in detecting cancer resistance to therapeutic agents.
Collapse
Affiliation(s)
- Chuanyun Li
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China
| | - Tong Zhou
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Chen
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China.,Beijing Institute of Hepatology, Beijing, China
| | - Rong Li
- Chengde Medical University, Chengde, China
| | - Huan Chen
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China
| | - Shumin Luo
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China.,Beijing Institute of Hepatology, Beijing, China
| | - Dexi Chen
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China.,Beijing Institute of Hepatology, Beijing, China
| | - Cao Cai
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China.
| | - Weihua Li
- Fengtai District, YouAn Hospital, Capital Medical University, NO. 8, Xitoutiao, Youanmen wai, Beijing, China. .,Beijing Institute of Hepatology, Beijing, China.
| |
Collapse
|
28
|
Kakizaki M, Yamamoto Y, Nakayama S, Kameda K, Nagashima E, Ito M, Suyama T, Matsuzaki Y, Chiba T, Sumiyoshi H, Inagaki Y, Kotani A. Human hepatocyte-derived extracellular vesicles attenuate the carbon tetrachloride-induced acute liver injury in mice. Cell Death Dis 2021; 12:1010. [PMID: 34707093 PMCID: PMC8551237 DOI: 10.1038/s41419-021-04204-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/25/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022]
Abstract
Acute liver injury (ALI) induced by chemicals or viruses can progress rapidly to acute liver failure (ALF), often resulting in death of patients without liver transplantation. Since liver transplantation is limited due to a paucity of donors, expensive surgical costs, and severe immune rejection, novel therapies are required to treat liver injury. Extracellular vesicles (EVs) are used for cellular communication, carrying RNAs, proteins, and lipids and delivering them intercellularly after being endocytosed by target cells. Recently, it was reported that EVs secreted from human hepatocytes have an ability to modulate the immune responses; however, these roles of EVs secreted from human hepatocytes were studied only with in vitro experiments. In the present study, we evidenced that EVs secreted from human hepatocytes attenuated the CCL4-induced ALI by inhibiting the recruitment of monocytes through downregulation of chemokine receptor in the bone marrow and recruitment of neutrophils through the reduction of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2 expression levels in the liver.
Collapse
Affiliation(s)
- Masatoshi Kakizaki
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Yuichiro Yamamoto
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Shunya Nakayama
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Kazuaki Kameda
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan ,grid.415020.20000 0004 0467 0255Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, 330-8503 Japan
| | - Etsuko Nagashima
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Masatoshi Ito
- grid.265061.60000 0001 1516 6626Support Center for Medical Research and Education, Tokai University School of Medicine, Kanagawa, 259-1193 Japan
| | - Takashi Suyama
- grid.411621.10000 0000 8661 1590Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane Japan
| | - Yumi Matsuzaki
- grid.411621.10000 0000 8661 1590Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane Japan
| | - Tetsuhiro Chiba
- grid.136304.30000 0004 0370 1101Department of Gastroenterology, Chiba University, Graduate School of Medicine, Inohana, Chuo-ku, Chiba, 260-8670 Japan
| | - Hideaki Sumiyoshi
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,grid.265061.60000 0001 1516 6626Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193 Japan ,Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Yutaka Inagaki
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,grid.265061.60000 0001 1516 6626Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193 Japan ,Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Ai Kotani
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| |
Collapse
|
29
|
miRNA and long non-coding RNA transcriptional expression in hepatocellular carcinoma cell line-secreted extracellular vesicles. Clin Exp Med 2021; 22:245-255. [PMID: 34319456 DOI: 10.1007/s10238-021-00744-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022]
Abstract
Extracellular vesicles (EVs) are membrane-released vesicles acting as transporters of proteins, lipids and short/long non-coding RNA (miRNAs and lncRNAs). They are released by normal and pathological cells, including hepatocellular carcinoma (HCC). To date, studies focused on miRNAs and lncRNAs contained in EVs derived from HCC are limited. Our aim was to analyze the transcriptional profile of potential regulating miRNAs and lncRNAs in EVs secreted by HCC tumor cell line (HepG2, n = 6), and from a non-tumorigenic hepatocyte cell line (WRL68, n = 6), to compare their differential expression profile and to identify novel molecular diagnostic markers of HCC. EVs were isolated from the conditioned medium, through differential centrifugations. The expression profile of miRNAs (miR-23a, miR-16-2, miR-181a, miR-373, miR-205, miR-27a, miR-1323, and miR-532) and lncRNAs (HULC, HOTAIR, XIST, MALAT-1, GAS-5, H19) was performed in Real-time PCR, and their transcript was found both in HepG2 and WRL68 EVs. Lower miR-181a, miR-205 and miR-1323 expression were detected in EVs secreted by HepG2 compared to WRL68, while an opposite trend was observed for miR-23a, miR-16-2, miR-373, miR-27a, and miR-532. Several significant correlations were found between miRNA and lncRNA. An in silico analysis was also performed. The results obtained could identified them as new potential diagnostic and prognostic biomarkers of HCC.
Collapse
|
30
|
Kim HS, Kim JS, Park NR, Nam H, Sung PS, Bae SH, Choi JY, Yoon SK, Hur W, Jang JW. Exosomal miR-125b Exerts Anti-Metastatic Properties and Predicts Early Metastasis of Hepatocellular Carcinoma. Front Oncol 2021; 11:637247. [PMID: 34386414 PMCID: PMC8354570 DOI: 10.3389/fonc.2021.637247] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Background & Aims Cancer metastasis is responsible for the majority of cancer-related deaths. Exosomal miRNAs have emerged as promising biomarkers for cancer, serving as signaling molecules that can regulate tumor growth and metastasis. This study examined circulating exosomal miRNAs that could predict hepatocellular carcinoma (HCC) metastasis. Methods Exosomal miRNA was measured by quantitative real-time PCR (qRT-PCR) in a large set of patients (n = 284). To investigate the role of exosomal miRNA in HCC, we performed a series of in vitro tests, such as exosome labeling, qRT-PCR, reverse transcription PCR, wound healing assay, transwell assay, and Western blot assay. Results Exosomal miR-125b was drastically downregulated in HCC patients with metastasis than in those without metastasis. In vitro, we observed the uptake of miR-125b by exosome in recipient cells. Exosome-mediated miR-125b significantly inhibited migration and invasion abilities and downregulated the mRNA expressions of MMP-2, MMP-9, and MMP-14 in recipient cells via intercellular communication. Further investigation revealed that miR-125b suppressed SMAD2 protein expression in recipient cells by binding to its 3' untranslated regions. Exosome-mediated miR-125b transfer also disrupted TGF-β1-induced epithelial-mesenchymal transition and TGF-β1/SMAD signaling pathway in recipient cells by leading to a decrease of SMAD2 protein expression. Moreover, exosomal miR-125b was downregulated after metastasis compared with that at baseline in patients with serial measurements before and after metastasis. Conclusions The results imply that exosome-mediated miR-125b exerts anti-metastatic properties in HCC. These findings highlight that circulating exosomal miR-125b might represent a reliable biomarker with diagnostic and therapeutic implications for extrahepatic metastasis from HCC.
Collapse
Affiliation(s)
- Hye Seon Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin Seoub Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Na Ri Park
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Heechul Nam
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Pil Soo Sung
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Wonhee Hur
- Division of Chronic Viral Disease, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Chungbuk, South Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
31
|
The two facets of gp130 signalling in liver tumorigenesis. Semin Immunopathol 2021; 43:609-624. [PMID: 34047814 PMCID: PMC8443519 DOI: 10.1007/s00281-021-00861-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
The liver is a vital organ with multiple functions and a large regenerative capacity. Tumours of the liver are the second most frequently cause of cancer-related death and develop in chronically inflamed livers. IL-6-type cytokines are mediators of inflammation and almost all members signal via the receptor subunit gp130 and the downstream signalling molecule STAT3. We here summarize current knowledge on how gp130 signalling and STAT3 in tumour cells and cells of the tumour micro-environment drives hepatic tumorigenesis. We furthermore discuss very recent findings describing also anti-tumorigenic roles of gp130/STAT3 and important considerations for therapeutic interventions.
Collapse
|
32
|
27-Hydroxycholesterol is a specific factor in the neoplastic microenvironment of HCC that causes MDR via GRP75 regulation of the redox balance and metabolic reprogramming. Cell Biol Toxicol 2021; 38:311-324. [PMID: 33880675 DOI: 10.1007/s10565-021-09607-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/14/2021] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Due to the tissue specificity of the liver, long-term exposure to a high concentration of 27-hydroxycholesterol (27HC) is a special characteristic of the tumour microenvironment in hepatocellular carcinoma (HCC). However, what occurs after HCC cells are long-term exposure to 27HC and the molecular mechanisms involved remain largely unexamined. METHODS A long-term 27HC-treated HepG2 cell line and the xenografts in nude mice were used as experimental models. Molecular mechanisms were investigated using bioinformatics analysis and molecular biological experiments. RESULTS Here, we found that by inducing an increase in oxidative stress signalling, 27HC activated glucose-regulated protein 75 (GRP75). On the one hand, GRP75 resulted in a change in the redox balance by regulating ROS generation and antioxidant system activity via affecting MMP, NRF2, HO-1, and NQO1 levels. On the other hand, GRP75 modified the metabolic reprogramming process by regulating key factors (HIF-1α, p-Akt, and c-myc) and glucose uptake, facilitating HCC cell growth in the inhospitable microenvironment. These two factors caused HCC cells to resist 27HC-induced cytotoxicity and attain multidrug resistance (MDR). CONCLUSIONS Our present study not only identified 27HC, a characteristic component of the neoplastic microenvironment of HCC that causes MDR via GRP75 to regulate the redox balance and metabolic reprogramming, but also revealed that targeted intervention by the "switch"-like molecule GRP75 could reverse the effect of 27HC from cancer promotion to cytotoxicity in HCC, suggesting a new strategy for specific intervention of HCC.
Collapse
|
33
|
Wang Y, Gao R, Li J, Tang S, Li S, Tong Q, Li S. Downregulation of hsa_circ_0074854 Suppresses the Migration and Invasion in Hepatocellular Carcinoma via Interacting with HuR and via Suppressing Exosomes-Mediated Macrophage M2 Polarization. Int J Nanomedicine 2021; 16:2803-2818. [PMID: 33880025 PMCID: PMC8052130 DOI: 10.2147/ijn.s284560] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have been identified as key factors in the development of hepatocellular carcinoma (HCC). However, the role and potential molecular mechanism of circRNAs in HCC remain largely unclear. In addition, exosomes are known as important messengers of the cross-talk between tumor cells and immune cells, while the role of extracellular circRNAs in the cell-to-cell communication of tumor cells and immune cells remains not unclear. METHODS The level of hsa_circ_0074854 in HCC cell lines and HCC cell-derived exosomes was assessed using RT-qPCR assay. In addition, CCK-8 and transwell assays were used to determine the viability, migration and invasion of HCC cells. RESULTS Hsa_circ_0074854 expression was upregulated in HCC tissues and HCC cell lines. Additionally, hsa_circ_0074854 knockdown was found to inhibit HCC growth in vitro and in vivo. Mechanistically, hsa_circ_0074854 knockdown inhibited the migration and invasion of HCC cells via interacting with human antigen R (HuR) to reduce its stability. Furthermore, hsa_circ_0074854 can be transferred from HCC cells to macrophages via exosomes. Exosomes with downregulated hsa_circ_0074854 suppressed macrophage M2 polarization, which in turn suppressing migration and invasion of HCC cells both in vitro and in vivo. CONCLUSION Downregulation of hsa_circ_0074854 suppresses the migration and invasion in hepatocellular carcinoma via interacting with HuR and via suppressing exosomes-mediated macrophage M2 polarization. Collectively, these findings may help to understand the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Rongfen Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People’s Republic of China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, Hubei, People’s Republic of China
| | - Shaotao Tang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Shuai Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Shiwang Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| |
Collapse
|
34
|
New Insights into the Role of miR-29a in Hepatocellular Carcinoma: Implications in Mechanisms and Theragnostics. J Pers Med 2021; 11:jpm11030219. [PMID: 33803804 PMCID: PMC8003318 DOI: 10.3390/jpm11030219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most lethal human cancer globally. For advanced HCC, curable plan for advanced HCC is yet to be established, and the prognosis remains poor. The detail mechanisms underlying the progression of HCC tumorigenicity and the corruption of tumor microenvironment (TME) is complex and inconclusive. A growing body of studies demonstrate microRNAs (miRs) are important regulators in the tumorigenicity and TME development. Notably, mounting evidences indicate miR-29a play a crucial role in exerting hepatoprotective effect on various types of stress and involved in the progression of HCC, which elucidates their potential theragnostic implications. In this review, we reviewed the advanced insights into the detail mechanisms by which miR-29a dictates carcinogenesis, epigenetic program, and metabolic adaptation, and implicated in the sponging activity of competitive endogenous RNAs (ceRNA) and the TME components in the scenario of HCC. Furthermore, we highlighted its clinical significance in diagnosis and prognosis, as well as the emerging therapeutics centered on the activation of miR-29a.
Collapse
|
35
|
Chen W, Mao Y, Liu C, Wu H, Chen S. Exosome in Hepatocellular Carcinoma: an update. J Cancer 2021; 12:2526-2536. [PMID: 33854614 PMCID: PMC8040701 DOI: 10.7150/jca.54566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor in the digestive tract with limited therapeutic choices. Intercellular communication among cancer cells and their microenvironment is crucial to disease progression. Exosomes are extracellular vesicles secreted by multiple types of cells into the extracellular space, which contain a variety of active components of secretory cells, including lipids, proteins, RNA and DNA. This vesicle structure involves in the exchange of materials and information between cells and plays an important role in the development of many diseases. Studies have shown that exosomes participate in the communication between HCC cells and non-HCC cells and regulate the occurrence and development of hepatocellular carcinoma. Therefore, exosomes may be specific biomarkers for early diagnosis and metastasis of HCC, which are also potential targets for the treatment of HCC. This review summarizes the characteristic, types and biological functions of exosomes and discusses their research progress and application prospects in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Wei Chen
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yinqi Mao
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chenbin Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Han Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuying Chen
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
36
|
Zhao J, Li H, Zhao S, Wang E, Zhu J, Feng D, Zhu Y, Dou W, Fan Q, Hu J, Jia L, Liu L. Epigenetic silencing of miR-144/451a cluster contributes to HCC progression via paracrine HGF/MIF-mediated TAM remodeling. Mol Cancer 2021; 20:46. [PMID: 33658044 PMCID: PMC7927270 DOI: 10.1186/s12943-021-01343-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is among the malignancies with the highest mortality. The key regulators and their interactive network in HCC pathogenesis remain unclear. Along with genetic mutations, aberrant epigenetic paradigms, including deregulated microRNAs (miRNAs), exert profound impacts on hepatocyte transformation and tumor microenvironment remodeling; however, the underlying mechanisms are largely uncharacterized. METHODS We performed RNA sequencing on HCC specimens and bioinformatic analyses to identify tumor-associated miRNAs. The miRNA functional targets and their effects on tumor-infiltrating immune cells were investigated. The upstream events, particularly the epigenetic mechanisms responsible for miRNA deregulation in HCC, were explored. RESULTS The miR-144/miR-451a cluster was downregulated in HCC and predicted a better HCC patient prognosis. These miRNAs promoted macrophage M1 polarization and antitumor activity by targeting hepatocyte growth factor (HGF) and macrophage migration inhibitory factor (MIF). The miR-144/miR-451a cluster and EZH2, the catalytic subunit of polycomb repressive complex (PRC2), formed a feedback circuit in which miR-144 targeted EZH2 and PRC2 epigenetically repressed the miRNA genes via histone H3K27 methylation of the promoter. The miRNA cluster was coordinately silenced by distal enhancer hypermethylation, disrupting chromatin loop formation and enhancer-promoter interactions. Clinical examinations indicated that methylation of this chromatin region is a potential HCC biomarker. CONCLUSIONS Our study revealed novel mechanisms underlying miR-144/miR-451a cluster deregulation and the crosstalk between malignant cells and tumor-associated macrophages (TAMs) in HCC, providing new insights into HCC pathogenesis and diagnostic strategies.
Collapse
Affiliation(s)
- Junlong Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Development Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Huichen Li
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fourth Military Medical University, No.169 Changlexi Road, Xi'an, 710032, China
| | - Shoujie Zhao
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Enxin Wang
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, China
| | - Jun Zhu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fourth Military Medical University, No.169 Changlexi Road, Xi'an, 710032, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yejing Zhu
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Weijia Dou
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, China
| | - Qingling Fan
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, China
| | - Jie Hu
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fourth Military Medical University, No.169 Changlexi Road, Xi'an, 710032, China.
| | - Lei Liu
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, No.569 Xinsi Road, Xi'an, 710038, China. .,Department of Cell Biology, Fourth Military Medical University, No.169 Changlexi Road, Xi'an, 710032, China.
| |
Collapse
|
37
|
Bernardi S, Farina M. Exosomes and Extracellular Vesicles in Myeloid Neoplasia: The Multiple and Complex Roles Played by These " Magic Bullets". BIOLOGY 2021; 10:biology10020105. [PMID: 33540594 PMCID: PMC7912829 DOI: 10.3390/biology10020105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Extracellular vesicles (EVs) are released by the majority of cell types and can be isolated from both cell cultures and body fluids. They are involved in cell-to-cell communication and may shuttle different messages (RNA, DNA, and proteins). These messages are known to influence the microenvironment of cells and their behavior. In recent years, some evidence about the involvement of EVs and exosomes, an EV subgroup, in immunomodulation, the transfer of disease markers, and the treatment of myeloid malignancies have been reported. Little is known about these vesicles in this particular setting of hematologic neoplasia; here, we summarize and critically review the available results, aiming to encourage further investigations. Abstract Extracellular vesicles (exosomes, in particular) are essential in multicellular organisms because they mediate cell-to-cell communication via the transfer of secreted molecules. They are able to shuttle different cargo, from nucleic acids to proteins. The role of exosomes has been widely investigated in solid tumors, which gave us surprising results about their potential involvement in pathogenesis and created an opening for liquid biopsies. Less is known about exosomes in oncohematology, particularly concerning the malignancies deriving from myeloid lineage. In this review, we aim to present an overview of immunomodulation and the microenvironment alteration mediated by exosomes released by malicious myeloid cells. Afterwards, we review the studies reporting the use of exosomes as disease biomarkers and their influence in response to treatment, together with the recent experiences that have focused on the use of exosomes as therapeutic tools. The further development of new technologies and the increased knowledge of biological (exosomes) and clinical (myeloid neoplasia) aspects are expected to change the future approaches to these malignancies.
Collapse
Affiliation(s)
- Simona Bernardi
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- Correspondence: or ; Tel.: +39-0303998464
| | - Mirko Farina
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
38
|
Human Hepatocellular Carcinoma (HCC). Cancers (Basel) 2020; 12:cancers12123739. [PMID: 33322652 PMCID: PMC7763211 DOI: 10.3390/cancers12123739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and accounts for 8 [...].
Collapse
|
39
|
Zheng Z, Wen Y, Nie K, Tang S, Chen X, Lan S, Pan J, Jiang K, Jiang X, Liu P, Yan Y, Liu F, Liu Y, Li P. Construction of a 13-microRNA-based signature and prognostic nomogram for predicting overall survival in patients with hepatocellular carcinoma. Hepatol Res 2020; 50:1151-1163. [PMID: 32567216 DOI: 10.1111/hepr.13538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022]
Abstract
AIM Hepatocellular carcinoma (HCC) is a common malignancy associated with a poor prognosis due to difficulties in reliably estimating overall survival (OS). MicroRNAs (miRNAs) play critical roles in HCC initiation, progression, and metastasis and are highly correlated with patient prognosis. Thus, miRNA-based risk signatures and nomograms are urgently required for predicting OS in patients with HCC. METHODS We constructed a 13-miRNA-based signature and prognostic nomogram using 408 HCC samples and 58 normal tissues with miRNA sequencing data and clinical data from 323 patients downloaded from The Cancer Genome Atlas. A total of 195 patients were assigned as the internal validation cohort for verification and testing. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis was applied to investigate pathway enrichment for the signature. RESULTS We identified and validated a 13-miRNA risk signature highly associating with the OS of HCC patients. The signature showed good performances by calculating C-index, area under the curve, and calibration curves. After verification and testing using an internal validation cohort, the results yielded a miRNA-based signature and a prognostic nomogram with reliable predictive accuracy. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis indicated that various genes and multiple pathways were closely related to the mechanisms of HCC proliferation and metastasis. CONCLUSION We successfully identified a 13-miRNA-based signature and prognostic nomogram that are capable of predicting OS in patients with HCC.
Collapse
Affiliation(s)
- Zhihua Zheng
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yi Wen
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Kechao Nie
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Shuting Tang
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Xu Chen
- Department of Gastroenterology, The first Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Shaoyang Lan
- Department of Gastroenterology, The first Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jinglin Pan
- Department of Gastroenterology, The Hainan Hospital of Chinese Medicine, Haikou, 570100, China
| | - Kailin Jiang
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Xiaotao Jiang
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Peng Liu
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yanhua Yan
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Fengbin Liu
- Department of Gastroenterology, The first Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yufeng Liu
- Department of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Peiwu Li
- Department of Gastroenterology, The first Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| |
Collapse
|
40
|
Azparren-Angulo M, Royo F, Gonzalez E, Liebana M, Brotons B, Berganza J, Goñi-de-Cerio F, Manicardi N, Abad-Jordà L, Gracia-Sancho J, Falcon-Perez JM. Extracellular vesicles in hepatology: Physiological role, involvement in pathogenesis, and therapeutic opportunities. Pharmacol Ther 2020; 218:107683. [PMID: 32961265 DOI: 10.1016/j.pharmthera.2020.107683] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Since the first descriptions of hepatocyte-released exosome-like vesicles in 2008, the number of publications describing Extracellular Vesicles (EVs) released by liver cells in the context of hepatic physiology and pathology has grown exponentially. This growing interest highlights both the importance that cell-to-cell communication has in the organization of multicellular organisms from a physiological point of view, as well as the opportunity that these circulating organelles offer in diagnostics and therapeutics. In the present review, we summarize systematically and comprehensively the myriad of works that appeared in the last decade and lighted the discussion about the best opportunities for using EVs in liver disease therapeutics.
Collapse
Affiliation(s)
- Maria Azparren-Angulo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Esperanza Gonzalez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Marc Liebana
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Bruno Brotons
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Jesús Berganza
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Felipe Goñi-de-Cerio
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Nicoló Manicardi
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Laia Abad-Jordà
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain; Hepatology, Department of Biomedical Research, Inselspital & University of Bern, Switzerland
| | - Juan M Falcon-Perez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48015, Spain.
| |
Collapse
|
41
|
Cavallari C, Camussi G, Brizzi MF. Extracellular Vesicles in the Tumour Microenvironment: Eclectic Supervisors. Int J Mol Sci 2020; 21:E6768. [PMID: 32942702 PMCID: PMC7555174 DOI: 10.3390/ijms21186768] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
The tumour microenvironment (TME) plays a crucial role in the regulation of cell survival and growth by providing inhibitory or stimulatory signals. Extracellular vesicles (EV) represent one of the most relevant cell-to-cell communication mechanism among cells within the TME. Moreover, EV contribute to the crosstalk among cancerous, immune, endothelial, and stromal cells to establish TME diversity. EV contain proteins, mRNAs and miRNAs, which can be locally delivered in the TME and/or transferred to remote sites to dictate tumour behaviour. EV in the TME impact on cancer cell proliferation, invasion, metastasis, immune-escape, pre-metastatic niche formation and the stimulation of angiogenesis. Moreover, EV can boost or inhibit tumours depending on the TME conditions and their cell of origin. Therefore, to move towards the identification of new targets and the development of a novel generation of EV-based targeting approaches to gain insight into EV mechanism of action in the TME would be of particular relevance. The aim here is to provide an overview of the current knowledge of EV released from different TME cellular components and their role in driving TME diversity. Moreover, recent proposed engineering approaches to targeting cells in the TME via EV are discussed.
Collapse
Affiliation(s)
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | | |
Collapse
|