151
|
Bu H, Song Q, Zhang J, Wei Y, Liu B. Development of a Novel KCNN4-Related ceRNA Network and Prognostic Model for Renal Clear Cell Carcinoma. Anal Cell Pathol (Amst) 2023; 2023:2533992. [PMID: 39282155 PMCID: PMC11401688 DOI: 10.1155/2023/2533992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 09/18/2024] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) accounts for more than 80% of renal cell carcinomas. Yet, it has not been fully understood about the derivation and progression of the tumor, as well as the long-term benefits from multimodality therapy. Therefore, reliable and applicable molecular markers are urgently needed for the prediction of diagnosis and prognosis of ccRCC patients. Methods Genetic and clinical information of 533 ccRCC patients from The Cancer Genome Atlas database was collected for comprehensive bioinformatic analyses. UALCAN was used to detect gene expression in paired tumor samples. Two data sets from Gene Expression Omnibus database were analyzed to identify differentially expressed genes (DEGs), and Gene Set Enrichment Analysis was applied for the functional enrichment of DEGs. Tumor Immune Single Cell Hub and Tumor IMmune Estimation Resource databases were separately used for analyses of single-immune cell and immune cell infiltration. Encyclopedia of RNA Interactomes database was explored to predict targeted microRNAs (miRNAs) and corresponding long non-coding RNAs (lncRNAs). Cox regression analysis was performed for the construction of risk signature and prognosis model. Finally, quantitative real-time polymerase chain reaction and western blot were conducted for KCNN4 expression detection in cell lines and clinical samples. Small interfering RNA was employed to knock down KCNN4, and corresponding functional experiments were conducted on ccRCC cells as well. Results KCNN4 showed elevated expression in tumors and prominent clinical correlation in ccRCC. In total, 41 KCNN4-related genes were enriched, and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses showed they were intimately related to immune-related signaling pathways. Spearman's analysis revealed the significantly positive correlation of KCNN4 with immune cell infiltration. By integrating hub miRNA-let-7e-5p and four critical lncRNA, a competitive endogenous RNA network-based risk signature was constructed. The prognosis model derived from it showed considerable predictive value for survival of ccRCC patients. Finally, in vitro experiments confirmed the remarkable tumor-promoting role of KCNN4 in ccRCC cells. Conclusion KCNN4 significantly affected the immune status of tumor microenvironment and immunotherapy elements, through which it promoted tumor progression in ccRCC, and it could be a potential biomarker for prognosis and immunotherapy effects of ccRCC patients.
Collapse
Affiliation(s)
- Hengtao Bu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiang Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiexiu Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuang Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Bianjiang Liu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
152
|
Ciafrè SA, Russo M, Michienzi A, Galardi S. Long Noncoding RNAs and Cancer Stem Cells: Dangerous Liaisons Managing Cancer. Int J Mol Sci 2023; 24:ijms24031828. [PMID: 36768150 PMCID: PMC9915130 DOI: 10.3390/ijms24031828] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Decades of research have investigated the mechanisms that lead to the origin of cancer, striving to identify tumor-initiating cells. These cells, also known as cancer stem cells, are characterized by the ability to self-renew, to give rise to differentiated tumor populations, and on a larger scale, are deemed responsible not only for tumor initiation but also for recurrent tumors, often resistant to chemotherapy and radiotherapy. Long noncoding RNAs are RNA molecules longer than 200 nt, lacking the ability to code for proteins, with recognized roles as fine regulators of gene expression. They can exert these functions through a variety of mechanisms, acting at almost all steps of gene expression, from modulation of the epigenetic state of chromatin to modulation of protein stability. In all cases, lncRNAs do not work alone, but they always interact with other RNA molecules, either coding or non-coding, or with protein factors. In this review, we summarize the latest results obtained about the involvement of lncRNAs in the initiating cells of several types of tumors, and highlight the different mechanisms through which they work, while discussing how the modulation of a lncRNA can affect several aspects of tumor onset and progression.
Collapse
Affiliation(s)
- Silvia Anna Ciafrè
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
- Correspondence: (S.A.C.); (S.G.)
| | - Monia Russo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Alessandro Michienzi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Silvia Galardi
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- Correspondence: (S.A.C.); (S.G.)
| |
Collapse
|
153
|
Benito-Martín A, Jasiulionis MG, García-Silva S. Extracellular vesicles and melanoma: New perspectives on tumor microenvironment and metastasis. Front Cell Dev Biol 2023; 10:1061982. [PMID: 36704194 PMCID: PMC9871288 DOI: 10.3389/fcell.2022.1061982] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Secreted extracellular vesicles (EVs) are lipid bilayer particles without functional nucleus naturally released from cells which constitute an intercellular communication system. There is a broad spectrum of vesicles shed by cells based on their physical properties such as size (small EVs and large EVs), biogenesis, cargo and functions, which provide an increasingly heterogenous landscape. In addition, they are involved in multiple physiological and pathological processes. In cancer, EV release is opted by tumor cells as a beneficial process for tumor progression. Cutaneous melanoma is a cancer that originates from the melanocyte lineage and shows a favorable prognosis at early stages. However, when melanoma cells acquire invasive capacity, it constitutes the most aggressive and deadly skin cancer. In this context, extracellular vesicles have been shown their relevance in facilitating melanoma progression through the modulation of the microenvironment and metastatic spreading. In agreement with the melanosome secretory capacity of melanocytes, melanoma cells display an enhanced EV shedding activity that has contributed to the utility of melanoma models for unravelling EV cargo and functions within a cancer scenario. In this review, we provide an in-depth overview of the characteristics of melanoma-derived EVs and their role in melanoma progression highlighting key advances and remaining open questions in the field.
Collapse
Affiliation(s)
- Alberto Benito-Martín
- Facultad de Medicina, Unidad de Investigación Biomédica, Universidad Alfonso X El Sabio (UAX), Villanueva de la Cañada, Spain,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| | - Miriam Galvonas Jasiulionis
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| | - Susana García-Silva
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain,*Correspondence: Alberto Benito-Martín, ; Miriam Galvonas Jasiulionis, ; Susana García-Silva,
| |
Collapse
|
154
|
Tao X, Li Y, Fan S, Wu L, Xin J, Su Y, Xian X, Huang Y, Huang R, Fang W, Liu Z. Downregulation of Linc00173 increases BCL2 mRNA stability via the miR-1275/PROCA1/ZFP36L2 axis and induces acquired cisplatin resistance of lung adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2023; 42:12. [PMID: 36627670 PMCID: PMC9830831 DOI: 10.1186/s13046-022-02560-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/04/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND LINC00173 had been reported as a cisplatin (cis-diamminedichloroplatinum, DDP) chemotherapy-resistant inducer in small-cell lung cancer (SCLC) and lung squamous cell carcinoma (LUSC). This study aimed to display reverse data for LINC00173 as a DDP chemosensitivity-inducing factor in lung adenocarcinoma (LUAD). METHODS LINC00173 was screened from the Gene Expression Omnibus database (GSE43493). The expression level of LINC00173 in LUAD tissues and cell lines was detected using in situ hybridization and quantitative reverse transcription-polymerase chain reaction. Colony formation, cell viability, half-maximal inhibitory concentration, flow cytometry, and xenograft mouse model were used to evaluate the role of LINC00173 in the chemosensitivity of LUAD to DDP. The mechanism of LINC00173 in DDP resistance by mediating miR-1275/PROCA1/ZFP36L2 axis to impair BCL2 mRNA stability was applied, and co-immunoprecipitation, chromatin immunoprecipitation, RNA antisense purification, RNA immunoprecipitation, and luciferase reporter assays were performed. RESULTS LINC00173 downregulation in patients with DDP-resistant LUAD was correlated with poor prognosis. Further, LINC00173 expression was significantly reduced in DDP-resistant LUAD cells and DDP-treated human LUAD tissues. Suppressed LINC00173 expression in LUAD cells enhanced DDP chemoresistance in vivo and in vitro, while restored LINC00173 expression in DDP-resistant LUAD cells markedly regained chemosensitivity to DDP. Mechanistically, DDP-resistant LUAD cells activated PI3K/AKT signal and further elevated the c-Myc expression. The c-Myc, as an oncogenic transcriptional factor, bound to the promoter of LINC00173 and suppressed its expression. The reduced LINC00173 expression attenuated the adsorption of oncogenic miR-1275, downregulating the expression of miR-1275 target gene PROCA1. PROCA1 played a potential tumor-suppressive role inducing cell apoptosis and DDP chemosensitivity via recruiting ZFP36L2 to bind to the 3' untranslated region of BCL2, reducing the stability of BCL2 mRNA and thus activating the apoptotic signal. CONCLUSIONS This study demonstrated a novel and critical role of LINC00173. It was transcriptionally repressed by DDP-activated PI3K/AKT/c-Myc signal in LUAD, promoting DDP-acquired chemotherapeutic resistance by regulating miR-1275 to suppress PROCA1/ZFP36L2-induced BCL2 degradation, which led to apoptotic signal reduction. These data were not consistent with the previously described role of LINC00173 in SCLC or LUSC, which suggested that LINC00173 could play fine-tuned DDP resistance roles in different pathological subtypes of lung cancer. This study demonstrated that the diminished expression of LINC00173 might serve as an indicator of DDP-acquired resistance in LUAD.
Collapse
Affiliation(s)
- Xingyu Tao
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yang Li
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Songqing Fan
- grid.452708.c0000 0004 1803 0208The Second Xiangya Hospital of Central South University, Changsha, 410008 China
| | - Liyang Wu
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Jianyang Xin
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yun Su
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Xiaoyang Xian
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Yingying Huang
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Rongquan Huang
- grid.410737.60000 0000 8653 1072Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436 China
| | - Weiyi Fang
- grid.284723.80000 0000 8877 7471Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China ,grid.284723.80000 0000 8877 7471Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315 China
| | - Zhen Liu
- grid.284723.80000 0000 8877 7471Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315 China ,grid.410737.60000 0000 8653 1072Department of Pathology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| |
Collapse
|
155
|
CircRNA-PI4KB Induces Hepatic Lipid Deposition in Non-Alcoholic Fatty Liver Disease by Transporting miRNA-122 to Extra-Hepatocytes. Int J Mol Sci 2023; 24:ijms24021297. [PMID: 36674813 PMCID: PMC9863671 DOI: 10.3390/ijms24021297] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/01/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Ectopic fat deposition in the liver, known as non-alcoholic fatty liver disease (NAFLD), affects up to 30% of the worldwide population. miRNA-122, the most abundant liver-specific miRNA, protects hepatic steatosis and inhibits cholesterol and fatty acid synthesis in NAFLD. Previously, we have shown that compared with its expression in healthy controls, miRNA-122 decreased in the liver tissue but gradually increased in the serum of patients with non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, suggesting that miRNA-122 could have been transported to the serum. Here, we aimed to confirm and unravel the mechanism of transportation of miRNA-122 to extra-hepatocytes. Our findings showed a decrease in the intra-hepatocyte miRNA-122 and an increase in the extra-hepatocyte (medium level) miRNA-122, suggesting the miRNA-122 "escaped" from the intra-hepatocyte due to an increased extra-hepatocyte excretion. Using bioinformatics tools, we showed that miRNA-122 binds to circPI4KB, which was further validated by an RNA pull-down and luciferase reporter assay. The levels of circPI4KB in intra- and extra-hepatocytes corresponded to that of miRNA-122, and the overexpression of circPI4KB increased the miRNA-122 in extra-hepatocytes, consequently accomplishing a decreased protective role of miRNA-122 in inhibiting the lipid deposition. The present study provides a new explanation for the pathogenesis of the hepatic lipid deposition in NAFLD.
Collapse
|
156
|
Li B, Li W, Liu T, Zha L. Extracellular vesicles regulate the transmission of insulin resistance and redefine noncommunicable diseases. Front Mol Biosci 2023; 9:1024786. [PMID: 36699697 PMCID: PMC9868246 DOI: 10.3389/fmolb.2022.1024786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023] Open
Abstract
Noncommunicable diseases (NCDs), such as diabetes and related neurological disorders, are considered to not be directly transmissible from one person to another. However, NCDs may be transmissible in vivo through extracellular vesicles (EVs). A long-term high-fat diet (HFD) can induce a series of health issues like hyperlipidemia, type 2 diabetes mellitus (T2DM), and diabetic peripheral neuropathy (DPN) due to insulin resistance. Multiple molecular signaling changes can stimulate insulin resistance, especially blocking insulin signaling by increased insulin resistance inducer (phosphorylation of negative regulatory sites of insulin receptor substrate (IRS) proteins) and decreased tyrosine phosphorylation of insulin receptor substrate (phosphorylation of positive regulatory sites of IRS), thus leading to reduced phosphorylation of AKT enzymes. Current efforts to treat T2DM and prevent its complications mainly focus on improving insulin sensitivity, enhancing insulin secretion, or supplementing exogenous insulin based on a common assumption that insulin resistance is noncommunicable. However, insulin resistance is transmissible within multiple tissues or organs throughout the body. Exploring the regulatory roles of EVs in developing insulin resistance may provide novel and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Biao Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration Key Laboratory of Cosmetic Safety Evaluation, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Wan Li
- School of Physical Education, Hubei Minzu University, Enshi, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Longying Zha
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration Key Laboratory of Cosmetic Safety Evaluation, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
157
|
Pan Y, Lu X, Shu G, Cen J, Lu J, Zhou M, Huang K, Dong J, Li J, Lin H, Song H, Xu Q, Han H, Chen Z, Chen W, Luo J, Wei J, Zhang J. Extracellular Vesicle-Mediated Transfer of LncRNA IGFL2-AS1 Confers Sunitinib Resistance in Renal Cell Carcinoma. Cancer Res 2023; 83:103-116. [PMID: 36264173 PMCID: PMC9811158 DOI: 10.1158/0008-5472.can-21-3432] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/24/2022] [Accepted: 10/12/2022] [Indexed: 02/03/2023]
Abstract
Sunitinib resistance remains a serious challenge to the treatment of advanced and metastatic renal cell carcinoma (RCC), yet the mechanisms underlying this resistance are not fully understood. Here, we report that the long noncoding RNA IGFL2-AS1 is a driver of therapy resistance in RCC. IGFL2-AS1 was highly upregulated in sunitinib-resistant RCC cells and was associated with poor prognosis in patients with clear cell RCC (ccRCC) who received sunitinib therapy. IGFL2-AS1 enhanced TP53INP2 expression by competitively binding to hnRNPC, a multifunctional RNA-binding protein that posttranscriptionally suppresses TP53INP2 expression through alternative splicing. Upregulated TP53INP2 enhanced autophagy and ultimately led to sunitinib resistance. Meanwhile, IGFL2-AS1 was packaged into extracellular vesicles through hnRNPC, thus transmitting sunitinib resistance to other cells. N6-methyladenosine modification of IGFL2-AS1 was critical for its interaction with hnRNPC. In a patient-derived xenograft model of sunitinib-resistant ccRCC, injection of chitosan-solid lipid nanoparticles containing antisense oligonucleotide-IGFL2-AS1 successfully reversed sunitinib resistance. These findings indicate a novel molecular mechanism of sunitinib resistance in RCC and suggest that IGFL2-AS1 may serve as a prognostic indicator and potential therapeutic target to overcome resistance. SIGNIFICANCE Extracellular vesicle-packaged IGFL2-AS1 promotes sunitinib resistance by regulating TP53INP2-triggered autophagy, implicating this lncRNA as a potential therapeutic target in renal cell carcinoma.
Collapse
Affiliation(s)
- Yihui Pan
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuanxuan Lu
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| | - Guannan Shu
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junjie Cen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Lu
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mi Zhou
- Department of Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kangbo Huang
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiaqi Dong
- Department of Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaying Li
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haishan Lin
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongde Song
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quanhui Xu
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Han
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhenhua Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junhang Luo
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Corresponding Authors: Jiaxing Zhang, Department of Oncology, First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510000, China. E-mail: ; Jinhuan Wei, ; and Junhang Luo,
| | - Jinhuan Wei
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Corresponding Authors: Jiaxing Zhang, Department of Oncology, First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510000, China. E-mail: ; Jinhuan Wei, ; and Junhang Luo,
| | - Jiaxing Zhang
- Department of Oncology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Corresponding Authors: Jiaxing Zhang, Department of Oncology, First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou, Guangdong, 510000, China. E-mail: ; Jinhuan Wei, ; and Junhang Luo,
| |
Collapse
|
158
|
Wu R, Wang K, Gai Y, Li M, Wang J, Wang C, Zhang Y, Xiao Z, Jiang D, Gao Z, Xia X. Nanomedicine for renal cell carcinoma: imaging, treatment and beyond. J Nanobiotechnology 2023; 21:3. [PMID: 36597108 PMCID: PMC9809106 DOI: 10.1186/s12951-022-01761-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
The kidney is a vital organ responsible for maintaining homeostasis in the human body. However, renal cell carcinoma (RCC) is a common malignancy of the urinary system and represents a serious threat to human health. Although the overall survival of RCC has improved substantially with the development of cancer diagnosis and management, there are various reasons for treatment failure. Firstly, without any readily available biomarkers, timely diagnosis has been greatly hampered. Secondly, the imaging appearance also varies greatly, and its early detection often remains difficult. Thirdly, chemotherapy has been validated as unavailable for treating renal cancer in the clinic due to its intrinsic drug resistance. Concomitant with the progress of nanotechnological methods in pharmaceuticals, the management of kidney cancer has undergone a transformation in the recent decade. Nanotechnology has shown many advantages over widely used traditional methods, leading to broad biomedical applications ranging from drug delivery, prevention, diagnosis to treatment. This review focuses on nanotechnologies in RCC management and further discusses their biomedical translation with the aim of identifying the most promising nanomedicines for clinical needs. As our understanding of nanotechnologies continues to grow, more opportunities to improve the management of renal cancer are expected to emerge.
Collapse
Affiliation(s)
- Ruolin Wu
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Keshan Wang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yongkang Gai
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Mengting Li
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Jingjing Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Chenyang Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Yajing Zhang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zhiwei Xiao
- grid.413247.70000 0004 1808 0969Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dawei Jiang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zairong Gao
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Xiaotian Xia
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
159
|
Yan C, Wang P, Zhao C, Yin G, Meng X, Li L, Cai S, Meng B. Long Noncoding RNA MAGI2-AS3 Represses Cell Progression in Clear Cell Renal Cell Carcinoma by Modulating the miR-629-5p/PRDM16 Axis. Crit Rev Eukaryot Gene Expr 2023; 33:43-56. [PMID: 37602452 DOI: 10.1615/critreveukaryotgeneexpr.2023048338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The objective of this study was to determine the regulatory mechanism of MAGI2-AS3 in clear cell renal cell carcinoma (ccRCC), thereby supplying a new insight for ccRCC treatment. Expression data in TCGA-KIRC were obtained. Target gene lncRNA for research was determined using expression analysis and clinical analysis. lncRNA's downstream regulatory miRNA and mRNA were predicted by bioinformatics databases. ccRCC cell malignant phenotypes were detected via CCK-8, colony formation, Transwell migration, and invasion assays. The targeting relationship between genes was assessed through dual-luciferase reporter gene analysis. Kaplan-Meier (K-M) analysis was carried out to verify the effect of MAGI2-AS3, miR-629-5p, and PRDM16 on the survival rate of ccRCC patients. MAGI2-AS3 expression in ccRCC tissue and cells was shown to be markedly decreased and its expression to continuously decline with tumor progression. MAGI2-AS3 suppresses ccRCC proliferation and migration. Dual-luciferase assay showed that MAGI2-AS3 binds miR-629-5p and that miR-629-5p binds PRDM16. In addition, functional experiments showed that MAGI2-AS3 facilitates PRDM16 expression by repressing miR-629-5p expression, thereby suppressing ccRCC cell aggression. K-M analysis showed that upregulation of either MAGI2-AS3 or PRDM16 significantly improves ccRCC patient survival, while upregulation of miR-629-5p has no significant impact. MAGI2-AS3 sponges miR-629-5p to modulate PRDM16 to mediate ccRCC development. Meanwhile, the MAGI2-AS3/miR-629-5p/PRDM16 axis, as a regulatory pathway of ccRCC progression, may be a possible therapeutic target and prognostic indicator of ccRCC.
Collapse
Affiliation(s)
- Chengquan Yan
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Pengfei Wang
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Chaofei Zhao
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Guangwei Yin
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Xin Meng
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Lin Li
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Shengyong Cai
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| | - Bin Meng
- Department of Urology, Area 3, Tangshan Gongren Hospital, Tangshan City, Hebei Province, 063000, China
| |
Collapse
|
160
|
Tan M, Ge Y, Wang X, Wang Y, Liu Y, He F, Teng H. Extracellular Vesicles (EVs) in Tumor Diagnosis and Therapy. Technol Cancer Res Treat 2023; 22:15330338231171463. [PMID: 37122245 PMCID: PMC10134167 DOI: 10.1177/15330338231171463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
In recent years, extracellular vesicles (EVs) have gained significant attention due to their tremendous potential for clinical applications. EVs play a crucial role in various aspects, including tumorigenesis, drug resistance, immune escape, and reconstruction of the tumor microenvironment. Despite the growing interest in EVs, many questions still need to be addressed before they can be practically applied in clinical settings. This paper aims to review EVs' isolation methods, structure research, the roles of EVs in tumorigenesis and their mechanisms in multiple types of tumors, their potential application in drug delivery, and the expectations for their future in clinical research.
Collapse
Affiliation(s)
- Mingdian Tan
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Yizhi Ge
- The Affiliated Cancer Hospital of Nanjing Medical University (Jiangsu Cancer Hospital) and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiaogang Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yan Wang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Yi Liu
- School of Medicine, Asian Liver Center, Stanford, CA, USA
| | - Feng He
- Stanford University School of Medicine, Stanford, CA, USA
| | - Hongqi Teng
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
161
|
Importance and implications of exosomes in nephrology and urology. Pflugers Arch 2023; 475:153-166. [PMID: 36399151 PMCID: PMC9849294 DOI: 10.1007/s00424-022-02771-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Exosomes are extracellular vesicles that are formed by two invaginations of the plasma membrane and can be released by all eukaryotic cells. Because of their bioactive contents, including nucleic acids and proteins, exosomes can activate a variety of functions in their recipient cells. Due to the plethora of physiological and pathophysiological functions, exosomes have received a lot of attention from researchers over the past few years. However, there is still no consensus regarding isolation and characterization protocols of exosomes and their subtypes. This heterogeneity poses a lot of methodical challenges but also offers new clinical opportunities simultaneously. So far, exosome-based research is still mostly limited to preclinical experiments and early-stage clinical trials since the translation of experimental findings remains difficult. Exosomes could potentially play an important role as future diagnostic and prognostic agents and might also be part of the development of new treatment strategies. Therefore, they have previously been investigated in a variety of nephrological and urological conditions such as acute kidney injury or prostate cancer.
Collapse
|
162
|
Qian F, Huang Z, Zhong H, Lei Q, Ai Y, Xie Z, Zhang T, Jiang B, Zhu W, Sheng Y, Hu J, Brinker CJ. Analysis and Biomedical Applications of Functional Cargo in Extracellular Vesicles. ACS NANO 2022; 16:19980-20001. [PMID: 36475625 DOI: 10.1021/acsnano.2c11298] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) can facilitate essential communication among cells in a range of pathophysiological conditions including cancer metastasis and progression, immune regulation, and neuronal communication. EVs are membrane-enclosed vesicles generated through endocytic origin and contain many cellular components, including proteins, lipids, nucleic acids, and metabolites. Over the past few years, the intravesicular content of EVs has proven to be a valuable biomarker for disease diagnostics, involving cancer, cardiovascular diseases, and central nervous system diseases. This review aims to provide insight into EV biogenesis, composition, function, and isolation, present a comprehensive overview of emerging techniques for EV cargo analysis, highlighting their major technical features and limitations, and summarize the potential role of EV cargos as biomarkers in disease diagnostics. Further, progress and remaining challenges will be discussed for clinical diagnostic outlooks.
Collapse
Affiliation(s)
- Feiyang Qian
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Zena Huang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou 510970, P.R. China
| | - Hankang Zhong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Qi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yiru Ai
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Zihui Xie
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Tenghua Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Bowen Jiang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yan Sheng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Jiaming Hu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials and the Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
163
|
Ren SW, Cao GQ, Zhu QR, He MG, Wu F, Kong SM, Zhang ZY, Wang Q, Wang F. Exosomes derived from human umbilical cord mesenchymal stem cells promote osteogenesis through the AKT signaling pathway in postmenopausal osteoporosis. Aging (Albany NY) 2022; 14:10125-10136. [PMID: 36575048 PMCID: PMC9831744 DOI: 10.18632/aging.204453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022]
Abstract
Postmenopausal osteoporosis (PMO) is a relatively common disease characterized by low bone mass and microstructural changes of trabecular bone. The reduced bone strength is caused a variety of complications, including fragility fracture and sarcopenia. We used CCK-8 and EdU assays to evaluate cell proliferation rates. The osteogenesis effect was detected using ALP staining, alizarin red staining, and q-PCR. In vivo, the effects of exosomes derived from HUC-MSCs were evaluated using HE staining, IHC staining and Masson staining. In addition, we explored the mechanism of exosomes and found that the AKT signaling pathway played an important role in osteogenesis and cell proliferation. This paper mainly explored the function of exosomes derived from human umbilical cord mesenchymal stem cells (HUC-MSCs) and provided a new strategy for the treatment of postmenopausal osteoporosis. In conclusion, exogenous administration of exosomes can contribute to the treatment postmenopausal osteoporosis to a certain extent.
Collapse
Affiliation(s)
- Shi-Wei Ren
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, Shandong, China
| | - Guang-Qing Cao
- Department of Spine Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, Shandong, China
| | - Qing-Run Zhu
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, Shandong, China
| | - Min-Gang He
- Department of Gastrointestinal Surgery, Shandong Tumor Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Fang Wu
- Department of Health, 960th Hospital of PLA, Jinan 250031, Shandong, China
| | - Su-Mei Kong
- Department of Health, 960th Hospital of PLA, Jinan 250031, Shandong, China
| | - Zhao-Yan Zhang
- Department of Health, 960th Hospital of PLA, Jinan 250031, Shandong, China
| | - Qiang Wang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, Shandong, China
| | - Feng Wang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250014, Shandong, China
| |
Collapse
|
164
|
Jiang T, Zhu Z, Zhang J, Chen M, Chen S. Role of tumor-derived exosomes in metastasis, drug resistance and diagnosis of clear cell renal cell carcinoma. Front Oncol 2022; 12:1066288. [PMID: 36620603 PMCID: PMC9810999 DOI: 10.3389/fonc.2022.1066288] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Renal cancer is one of the most extensively studied human tumors today, with clear cell renal cell carcinoma accounting for approximately 80% of all cases. Despite recent advances in research on clear cell renal cell carcinoma, advanced distant metastasis of the disease, delay in diagnosis, as well as drug resistance remain major problems. In recent years, as an important mediator of material and information exchange between cells in the tumor microenvironment, exosomes have attracted widespread attention for their role in tumor development. It has been reported that tumor-derived exosomes may act as regulators and have an important effect on the metastasis, drug resistance formation, and providing targets for early diagnosis of clear cell renal cell carcinoma. Therefore, the extensive study of tumour-derived exosomes will provide a meaningful reference for the development of the diagnostic and therapeutic field of clear cell renal cell carcinoma. This article reviews the biological role and research progress of tumor-derived exosomes in different aspects of premetastatic niche formation, tumor angiogenesis, and epithelial-mesenchymal transition during the progression of clear cell renal cell carcinoma. In addition, the role of tumor-derived exosomes in the development of drug resistance in clear cell renal cell carcinoma is also addressed in this review. Furthermore, recent studies have found that cargoes of exosomes in serum and urine, for example, a series of miRNAs, have the potential to be biological markers of clear cell renal cell carcinoma and provide meaningful targets for early diagnosis and monitoring of tumors, which is also covered in this article.
Collapse
Affiliation(s)
- Tiancheng Jiang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Zepeng Zhu
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Jiawei Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| | - Shuqiu Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
- Department of Medical College, Southeast University, Nanjing, China
| |
Collapse
|
165
|
Singh A, Choudhury SD, Singh P, Singh VV, Singh SN, Sharma A. KCMF1 regulates autophagy and ion channels' function in renal cell carcinoma: a future therapeutic target. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04507-y. [PMID: 36515749 DOI: 10.1007/s00432-022-04507-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION In RCC, systematic procedures such as surgery, chemo-radiation therapy, and application of target-based inhibitors increase the risk of several comorbidities such as chronic kidney disease, hemorrhage, and cardiac arrest that may increase the mortality rate. Even though immune-based checkpoint inhibitor therapies have an overall good response rate, it is restricted to only 30-40% of patients. Hence, an in-depth study of tumor pathophysiology in RCC is needed to identify the new therapeutic target. In RCC, persisted hypoxia is an essential phenomenon for tumor growth and progression. KCMF1 is a newly identified ubiquitin ligase whose domain interacts with destabilized proteins and reprogrammed the ubiquitin coding for lysosome-mediated degradation and autophagy under hypoxic conditions/oxidative stress and maintaining cellular homeostasis. But in RCC, the functional role of KCMF1 remains undefined to date. METHOD We determined KCMF1 and its associated proteins RAD6 and UBR4 expression and their co-localization using confocal microscopy in tumor and non-tumor tissues samples. Further, immunofluorescence staining was performed to determine autophagy (LC3B, p62), hypoxia-inducible factor (HIF-1A) and ion channel markers (Kv1.3, KCNN4) in RCC patients (n-10). Inductively coupled plasma mass spectrophotometry (ICPMS) was performed to estimate the concentration of potassium (K+), sodium (Na+) and Zinc (zn2+) in tumor and non-tumor cells of RCC patients (n-20). Lastly, images were analyzed using ZEN3.1, and ImageJ software. RESULT AND CONCLUSION We observed a discrepancy in the formation of ubiquitin ligase, autophagosome via KCMF1, and ionic concentration in tumor cells, which might be one of the possible factors for cancer evolution. KCMF1-associated ubiquitin ligase system could be considered as a novel therapeutic target for RCC in the future.
Collapse
Affiliation(s)
- Ashu Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Saumitra Dey Choudhury
- Central Core Research Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhjot Singh
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Som Nath Singh
- Defence Institute of Physiology and Allied Sciences, New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
166
|
Pan Y, Tang H, Li Q, Chen G, Li D. Exosomes and their roles in the chemoresistance of pancreatic cancer. Cancer Med 2022; 11:4979-4988. [PMID: 35587712 PMCID: PMC9761084 DOI: 10.1002/cam4.4830] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 02/03/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal human malignancies worldwide. Due to the insidious onset and the rapid progression, most patients with PC are diagnosed at an advanced stage rendering them inoperable. Despite the development of multiple promising chemotherapeutic agents as recommended first-line treatment for PC, the therapeutic efficacy is largely limited by unwanted drug resistance. Recent studies have identified exosomes as essential mediators of intercellular communications during the occurrence of drug resistance. Understanding the underlying molecular mechanisms and complex signaling pathways of exosome-mediated drug resistance will contribute to the improvement of the design of new oncologic therapy regimens. This review focuses on the intrinsic connections between the chemoresistance of PC cells and exosomes in the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Yubin Pan
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Honglin Tang
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Qijun Li
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Guangpeng Chen
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
167
|
Yang Q, Xu J, Gu J, Shi H, Zhang J, Zhang J, Chen Z, Fang X, Zhu T, Zhang X. Extracellular Vesicles in Cancer Drug Resistance: Roles, Mechanisms, and Implications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201609. [PMID: 36253096 PMCID: PMC9731723 DOI: 10.1002/advs.202201609] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 09/10/2022] [Indexed: 06/16/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanosized vesicles that mediate cell-to-cell communication via transporting bioactive molecules and thus are critically involved in various physiological and pathological conditions. EVs contribute to different aspects of cancer progression, such as cancer growth, angiogenesis, metastasis, immune evasion, and drug resistance. EVs induce the resistance of cancer cells to chemotherapy, radiotherapy, targeted therapy, antiangiogenesis therapy, and immunotherapy by transferring specific cargos that affect drug efflux and regulate signaling pathways associated with epithelial-mesenchymal transition, autophagy, metabolism, and cancer stemness. In addition, EVs modulate the reciprocal interaction between cancer cells and noncancer cells in the tumor microenvironment (TME) to develop therapy resistance. EVs are detectable in many biofluids of cancer patients, and thus are regarded as novel biomarkers for monitoring therapy response and predicting prognosis. Moreover, EVs are suggested as promising targets and engineered as nanovehicles to deliver drugs for overcoming drug resistance in cancer therapy. In this review, the biological roles of EVs and their mechanisms of action in cancer drug resistance are summarized. The preclinical studies on using EVs in monitoring and overcoming cancer drug resistance are also discussed.
Collapse
Affiliation(s)
- Qiurong Yang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jing Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory MedicineNantong Tumor HospitalNantongJiangsu226361China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jiayin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| | - Jianye Zhang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical PharmacologySchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouGuangdong511436China
| | - Zhe‐Sheng Chen
- College of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Xinjian Fang
- Department of OncologyLianyungang Hospital Affiliated to Jiangsu UniversityLianyungangJiangsu222000China
| | - Taofeng Zhu
- Department of Pulmonary and Critical Care MedicineYixing Hospital affiliated to Jiangsu UniversityYixingJiangsu214200China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory MedicineSchool of MedicineJiangsu UniversityZhenjiangJiangsu212013China
| |
Collapse
|
168
|
Amendolare A, Marzano F, Petruzzella V, Vacca RA, Guerrini L, Pesole G, Sbisà E, Tullo A. The Underestimated Role of the p53 Pathway in Renal Cancer. Cancers (Basel) 2022; 14:cancers14235733. [PMID: 36497215 PMCID: PMC9736171 DOI: 10.3390/cancers14235733] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The TP53 tumor suppressor gene is known as the guardian of the genome, playing a pivotal role in controlling genome integrity, and its functions are lost in more than 50% of human tumors due to somatic mutations. This percentage rises to 90% if mutations and alterations in the genes that code for regulators of p53 stability and activity are taken into account. Renal cell carcinoma (RCC) is a clear example of cancer that despite having a wild-type p53 shows poor prognosis because of the high rate of resistance to radiotherapy or chemotherapy, which leads to recurrence, metastasis and death. Remarkably, the fact that p53 is poorly mutated does not mean that it is functionally active, and increasing experimental evidences have demonstrated this. Therefore, RCC represents an extraordinary example of the importance of p53 pathway alterations in therapy resistance. The search for novel molecular biomarkers involved in the pathways that regulate altered p53 in RCC is mandatory for improving early diagnosis, evaluating the prognosis and developing novel potential therapeutic targets for better RCC treatment.
Collapse
Affiliation(s)
- Alessandra Amendolare
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, 70121 Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Luisa Guerrini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Elisabetta Sbisà
- Institute of Biomedical Technologies, National Research Council—CNR, 70126 Bari, Italy
| | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
- Correspondence: ; Tel.: +39-0805929672
| |
Collapse
|
169
|
A Computationally Constructed lncRNA-Associated Competing Triplet Network in Clear Cell Renal Cell Carcinoma. DISEASE MARKERS 2022; 2022:8928282. [DOI: 10.1155/2022/8928282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/19/2022]
Abstract
Long noncoding RNAs (lncRNAs) are revealed to be involved in the tumorigenesis and progression of human malignancies mediated by microRNA (miRNA) via the competing endogenous RNA (ceRNA) mechanism, a newly proposed “RNA language.” However, the lncRNA-associated competing triplet (lncACT) network among ceRNA transcripts in clear cell renal cell carcinoma (ccRCC) is currently lacking. We carried out differential expression analysis to identify aberrantly expressed lncRNAs, miRNAs, and mRNAs by analyzing the RNA-seq data of 420 ccRCC tissues and 71 noncancerous kidney tissues obtained from The Cancer Genome Atlas (TCGA). Then, a ccRCC-specific ceRNA network was built using computational algorithms, including miRcode, TargetScan, miRanda, and miRTarBase. In total, 1491 dysregulated lncRNAs were found between normal renal tissues and ccRCC (fold
and false discovery
). A ceRNA network that comprised of 46 DElncRNAs, 11 DEmiRNAs, and 55 DEmRNAs was established by integrating the lncRNA/miRNA and miRNA/mRNA interactions into lncACTs. Several lncRNAs were identified to be significantly associated with clinical features of ccRCC patients. Notably, four key lncRNAs (TCL6, HOTTIP, HULC, and PCGEM1) were tightly correlated with both patients’ clinical characteristics and overall survival (log-rank
), indicating their potential important roles in ccRCC. HOTTIP may be a potential prognostic and therapeutic molecular marker for ccRCC patients. Collectively, our results provide a comprehensive view of the lncRNA-associated ceRNA regulatory network for a better understanding of the mechanisms and prognosis biomarkers for ccRCC.
Collapse
|
170
|
Liu T, Zhao H. Long Non-Coding RNAs: A Double-Edged Sword in Renal Cell Carcinoma Carcinogenesis. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1537.1549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
171
|
Hashemi M, Hajimazdarany S, Mohan CD, Mohammadi M, Rezaei S, Olyaee Y, Goldoost Y, Ghorbani A, Mirmazloomi SR, Gholinia N, Kakavand A, Salimimoghadam S, Ertas YN, Rangappa KS, Taheriazam A, Entezari M. Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance. Pharmacol Res 2022; 186:106535. [DOI: 10.1016/j.phrs.2022.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
|
172
|
Abdi E, Latifi-Navid S. Emerging long noncoding RNA polymorphisms as novel predictors of survival in cancer. Pathol Res Pract 2022; 239:154165. [DOI: 10.1016/j.prp.2022.154165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 11/09/2022]
|
173
|
Ebrahimi N, Faghihkhorasani F, Fakhr SS, Moghaddam PR, Yazdani E, Kheradmand Z, Rezaei-Tazangi F, Adelian S, Mobarak H, Hamblin MR, Aref AR. Tumor-derived exosomal non-coding RNAs as diagnostic biomarkers in cancer. Cell Mol Life Sci 2022; 79:572. [PMID: 36308630 PMCID: PMC11802992 DOI: 10.1007/s00018-022-04552-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 12/24/2022]
Abstract
Almost all clinical oncologists agree that the discovery of reliable, accessible, and non-invasive biomarkers is necessary to decrease cancer mortality. It is possible to employ reliable biomarkers to diagnose cancer in the early stages, predict the patient prognosis, follow up the response to treatment, and estimate the risk of disease recurrence with high sensitivity and specificity. Extracellular vesicles (EVs), especially exosomes, have been the focus of translational research to develop such biomarkers over the past decade. The abundance and distribution of exosomes in bodily fluids, including serum, saliva, and urine, as well as their ability to transport various biomolecules (nucleic acids, proteins, and lipids) derived from their parent cells, make exosomes reliable, accessible, and potent biomarkers for diagnosis and follow-up of solid and hematopoietic tumors. In addition, exosomes play a vital role in various cellular processes, including tumor progression, by participating in intercellular communication. Although these advantages underline the high potential of tumor-derived exosomes as diagnostic biomarkers, the lack of standardized effective methods for their isolation, identification, and precise characterization makes their application challenging in clinical settings. We discuss the importance of non-coding RNAs (ncRNAs) in cellular processes, and the role of tumor-derived exosomes containing ncRNAs as potential biomarkers in several types of cancer. In addition, the advantages and challenges of these studies for translation into clinical applications are covered.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Siavash Seifollahy Fakhr
- Division of Biotechnology, Faculty of Applied Ecology, Agricultural Sciences and Biotechnology, Campus, Hamar, Norway
| | - Parichehr Roozbahani Moghaddam
- Department of Molecular Genetics, Faculty of Science, Tonekabon Branch, Islamic Azad University, Tehran, Mazandaran, Iran
| | - Elnaz Yazdani
- Department of Biology, Faculty of Science, University of Isfahan, Isfahan, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Zahra Kheradmand
- Department of Agriculture, Islamic Azad University Maragheh Branch, Maragheh, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Samaneh Adelian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Halimeh Mobarak
- Clinical Pathologist, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Translational Medicine Group, Xsphera Biosciences, 6 Tide Street, Boston, MA, 02210, USA.
| |
Collapse
|
174
|
Yuan X, Jing Y, Guang M, Zhu J, Wang J, Wang Y, Zhang Y. GAS5 alleviates cisplatin drug resistance in oral squamous cell carcinoma by sponging miR-196a. J Int Med Res 2022; 50:3000605221132456. [PMID: 36310502 PMCID: PMC9619287 DOI: 10.1177/03000605221132456] [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] [Indexed: 11/06/2022] Open
Abstract
Objective The long non-coding RNA Growth-arrest-specific transcript 5 (GAS5) has been extensively linked with the ability of cancer cells to resist chemotherapeutic interventions. This prospective study aimed to investigate the role of GAS5 in oral squamous cell carcinoma (OSCC), which has been poorly characterized to date. Methods GAS5 and miR-196a expression levels were detected by quantitative real-time PCR analysis. Cisplatin (DDP) sensitivity and apoptosis levels were determined using Cell Counting Kit 8 and flow cytometry, respectively. Luciferase reporter and RNA immunoprecipitation assays were performed to confirm target miRNAs of GAS5. Results We found that GAS5 was expressed at low levels in DDP-resistant OSCC cell lines and tissues, and that GAS5 levels were intricately linked to the survival rates of OSCC patients. GAS5 overexpression led to the recovery of DDP sensitivity in CAL27/DDP cells. Additionally, in both DDP-resistant and -sensitive lines, GAS5 showed a cytoplasmic distribution and downregulated miR-196a in OSCC tissues. Exogenous transfection of miR-196a alleviated the effects of GAS5 on DDP sensitivity, confirming this as the mechanism of chemoresistance. Conclusions These findings may provide new targets for the treatment of chemotherapy-resistant OSCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ye Zhang
- Ye Zhang, Center of Stomatology, China-Japan Friendship Hospital, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
175
|
Ren W, Yuan Y, Peng J, Mutti L, Jiang X. The function and clinical implication of circular RNAs in lung cancer. Front Oncol 2022; 12:862602. [PMID: 36338714 PMCID: PMC9629004 DOI: 10.3389/fonc.2022.862602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Despite the recent advent of promising new targeted therapies, lung cancer diagnostic strategies still have difficulty in identifying the disease at an early stage. Therefore, the characterizations of more sensible and specific cancer biomarkers have become an important goal for clinicians. Circular RNAs are covalently close, endogenous RNAs without 5' end caps or 3'poly (A) tails and have been characterized by high stability, abundance, and conservation as well as display cell/tissue/developmental stage-specific expressions. Numerous studies have confirmed that circRNAs act as microRNA (miRNA) sponges, RNA-binding protein, and transcriptional regulators; some circRNAs even act as translation templates that participate in multiple pathophysiological processes. Growing evidence have confirmed that circRNAs are involved in the pathogenesis of lung cancers through the regulation of proliferation and invasion, cell cycle, autophagy, apoptosis, stemness, tumor microenvironment, and chemotherapy resistance. Moreover, circRNAs have emerged as potential biomarkers for lung cancer diagnosis and prognosis and targets for developing new treatments. In this review, we will summarize recent progresses in identifying the biogenesis, biological functions, potential mechanisms, and clinical applications of these molecules for lung cancer diagnosis, prognosis, and targeted therapy.
Collapse
Affiliation(s)
- Wenjun Ren
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yixiao Yuan
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Luciano Mutti
- The Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Xiulin Jiang
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
176
|
Xu D, Di K, Fan B, Wu J, Gu X, Sun Y, Khan A, Li P, Li Z. MicroRNAs in extracellular vesicles: Sorting mechanisms, diagnostic value, isolation, and detection technology. Front Bioeng Biotechnol 2022; 10:948959. [PMID: 36324901 PMCID: PMC9618890 DOI: 10.3389/fbioe.2022.948959] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of short, single-stranded, noncoding RNAs, with a length of about 18–22 nucleotides. Extracellular vesicles (EVs) are derived from cells and play a vital role in the development of diseases and can be used as biomarkers for liquid biopsy, as they are the carriers of miRNA. Existing studies have found that most of the functions of miRNA are mainly realized through intercellular transmission of EVs, which can protect and sort miRNAs. Meanwhile, detection sensitivity and specificity of EV-derived miRNA are higher than those of conventional serum biomarkers. In recent years, EVs have been expected to become a new marker for liquid biopsy. This review summarizes recent progress in several aspects of EVs, including sorting mechanisms, diagnostic value, and technology for isolation of EVs and detection of EV-derived miRNAs. In addition, the study reviews challenges and future research avenues in the field of EVs, providing a basis for the application of EV-derived miRNAs as a disease marker to be used in clinical diagnosis and even for the development of point-of-care testing (POCT) platforms.
Collapse
Affiliation(s)
- Dongjie Xu
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Kaili Di
- Department of Laboratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Boyue Fan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinrui Gu
- Department of Laboratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yifan Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education (Southeast University), Southeast University, Nanjing, China
| | - Peng Li
- College of Animal Science, Yangtze University, Jingzhou, China
- *Correspondence: Peng Li, ; Zhiyang Li,
| | - Zhiyang Li
- Department of Laboratory Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Peng Li, ; Zhiyang Li,
| |
Collapse
|
177
|
Cancer-associated fibroblast-specific lncRNA LINC01614 enhances glutamine uptake in lung adenocarcinoma. J Hematol Oncol 2022; 15:141. [PMID: 36209111 PMCID: PMC9548164 DOI: 10.1186/s13045-022-01359-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022] Open
Abstract
Background Besides featured glucose consumption, recent studies reveal that cancer cells might prefer “addicting” specific energy substrates from the tumor microenvironment (TME); however, the underlying mechanisms remain unclear. Methods Fibroblast-specific long noncoding RNAs were screened using RNA-seq data of our NJLCC cohort, TCGA, and CCLE datasets. The expression and package of LINC01614 into exosomes were identified using flow cytometric sorting, fluorescence in situ hybridization (FISH), and quantitative reverse transcription polymerase chain reaction (RT-PCR). The transfer and functional role of LINC01614 in lung adenocarcinoma (LUAD) and CAFs were investigated using 4-thiouracil-labeled RNA transfer and gain- and loss-of-function approaches. RNA pull-down, RNA immunoprecipitation, dual-luciferase assay, gene expression microarray, and bioinformatics analysis were performed to investigate the underlying mechanisms involved. Results We demonstrate that cancer-associated fibroblasts (CAFs) in LUAD primarily enhance the glutamine metabolism of cancer cells. A CAF-specific long noncoding RNA, LINC01614, packaged by CAF-derived exosomes, mediates the enhancement of glutamine uptake in LUAD cells. Mechanistically, LINC01614 directly interacts with ANXA2 and p65 to facilitate the activation of NF-κB, which leads to the upregulation of the glutamine transporters SLC38A2 and SLC7A5 and eventually enhances the glutamine influx of cancer cells. Reciprocally, tumor-derived proinflammatory cytokines upregulate LINC01614 in CAFs, constituting a feedforward loop between CAFs and cancer cells. Blocking exosome-transmitted LINC01614 inhibits glutamine addiction and LUAD growth in vivo. Clinically, LINC01614 expression in CAFs is associated with the glutamine influx and poor prognosis of patients with LUAD. Conclusion Our study highlights the therapeutic potential of targeting a CAF-specific lncRNA to inhibit glutamine utilization and cancer progression in LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01359-4.
Collapse
|
178
|
Construction and Characterization of n6-Methyladenosine-Related lncRNA Prognostic Signature and Immune Cell Infiltration in Kidney Renal Clear Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:7495183. [PMID: 36213821 PMCID: PMC9536954 DOI: 10.1155/2022/7495183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022]
Abstract
Background. Kidney renal clear cell carcinoma (KIRC) lacks effective prognostic biomarkers and the role and mechanism of N6-methyladenosine (m6A) modification of long noncoding RNAs (lncRNAs) in KIRC remain unclear. Methods. We extracted standard mRNA-sequencing and clinical data from the TCGA database. The prognostic risk model was obtained by Lasso regression and Cox regression. We randomly divided the samples into training and test sets, each taking half of the cases. Based on Lasso regression and Cox regression for training set, the prognostic risk signature was constructed; risk scores were calculated with the R package “glmnet.” Based on the median value of the prognostic risk score, risk scores were calculated for each patient and we divided all KIRC samples into high-risk and low-risk groups. Then, high- and low-risk subtypes were established and their prognosis, clinical features, and immune infiltration microenvironment were evaluated in test set and the entire sampled data set. The reliability of the prognostic model was confirmed by receiver operating characteristic curve analysis. Results. We found 28 prognostic m6A-related lncRNAs and established a m6A-related lncRNAs prognostic signature.
The signature showed a better predictive ability than other clinical indicators, including tumor node metastasis classification (TNM), histological, and pathological stages. In the high-risk group, M0 macrophages, CD8+ T cells, and regulatory T cells had significantly higher scores. Contrarily, in the low-risk group, activated dendritic cells, M1 macrophages, mast resting cells, and monocytes had significantly higher scores. In the high-risk group, LSECtin was overexpressed. In the low-risk group, PD-L1 was overexpressed. Moreover, high-risk patients may benefit more from AZ628. Conclusions. In conclusion, prognosis prediction of patients with KIRC and new insights for immunotherapy are provided by the m6A-related lncRNA prognostic signature.
Collapse
|
179
|
Hao X, Zhang Y, Shi X, Liu H, Zheng Z, Han G, Rong D, Zhang C, Tang W, Wang X. CircPAK1 promotes the progression of hepatocellular carcinoma via modulation of YAP nucleus localization by interacting with 14-3-3ζ. J Exp Clin Cancer Res 2022; 41:281. [PMID: 36131287 PMCID: PMC9494907 DOI: 10.1186/s13046-022-02494-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Circular RNA (circRNA), a new class of non-coding RNA, has obvious correlations with the occurrence and development of many diseases, including tumors. This study aimed to investigate the potential roles of circPAK1 in hepatocellular carcinoma (HCC).
Methods
High-throughput sequencing was performed on 3 pairs of HCC and matched normal tissues to determine the upregulated circRNAs. The expression level of circPAK1 was detected by qRT-PCR in HCC and paired with normal liver tissue samples. The effects of circPAK1 on proliferation, invasion, metastasis and apoptosis of HCC cells were evaluated by in vitro and in vivo experiments. We also constructed Chitosan/si-circPAK1 (CS/si-circPAK1) nanocomplexes using Chitosan material to evaluate its in vivo therapeutic effect on HCC. High-throughput sequencing, RNA-sequencing, RNA probe pull-down, RNA immunoprecipitation and Co-Immunoprecipitation assays were performed to explore the relationship between circPAK1, 14–3-3ζ, p-LATS1 and YAP. Exosomes isolated from lenvatinib-resistant HCC cell lines were used to evaluate the relationship between exosomal circPAK1 and lenvatinib resistance.
Results
CircPAK1, a novel circRNA, is highly expressed in HCC tumor tissues and cell lines as well as correlated with poor outcomes in HCC patients. Functionally, circPAK1 knockdown inhibited HCC cell proliferation, migration, invasion and angiogenesis while circPAK1 overexpression promoted HCC progression. The tumor-promoting phenotypes of circPAK1 on HCC were also confirmed by animal experiments. Importantly, the application of CS/si-circPAK1 nanocomplexes showed a better therapeutic effect on tumor growth and metastasis. Mechanistically, circPAK1 enhanced HCC progression by inactivating the Hippo signaling pathway, and this kind of inactivation is based on its competitively binding of 14–3-3 ζ with YAP, which weakens the recruitment and cytoplasmic fixation of 14–3-3 ζ to YAP, thus promoting YAP nucleus localization. Additionally, circPAK1 could be transported by exosomes from lenvatinib-resistant cells to sensitive cells and induce lenvatinib resistance of receipt cells.
Conclusion
CircPAK1 exerts its oncogenic function by competitively binding 14–3-3 ζ with YAP, thus promoting YAP nucleus localization, leading to the inactivation of a Hippo signaling pathway. Exosomal circPAK1 may drive resistance to lenvatinib, providing a potential therapeutic target for HCC patients.
Collapse
|
180
|
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long Non-coding RNAs (lncRNAs) signaling in Cancer Chemoresistance: From Prediction to Druggability. Drug Resist Updat 2022; 65:100866. [DOI: 10.1016/j.drup.2022.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022]
|
181
|
Zhang M, Wu L, Wang X, Chen J. lncKRT16P6 promotes tongue squamous cell carcinoma progression by sponging miR‑3180 and regulating GATAD2A expression. Int J Oncol 2022; 61:111. [PMID: 35904180 PMCID: PMC9374467 DOI: 10.3892/ijo.2022.5401] [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: 04/01/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022] Open
Abstract
Tongue squamous cell carcinoma (TSCC) is characterized by a poor prognosis and its 5‑year overall survival rate has not improved significantly. However, the precise molecular mechanisms underlying TSCC remain largely unknown. Through RNA screening, the present study identified a novel long noncoding RNA (lncRNA), keratin 16 pseudogene 6 (lncKRT16P6), which was upregulated in TSCC tissues and cell lines and associated with TSCC tumor stage and differentiation grade. Inhibition of lncKRT16P6 expression reduced TSCC cell migration, invasion and proliferation. lncKRT16P6 sponged microRNA (miR)‑3180 and upregulated GATA zinc finger domain containing 2A (GATAD2A) expression. miR‑3180 inhibition reversed the lncKRT16P6 depletion‑induced attenuation of TSCC malignancy and GATAD2A depletion reversed the miR‑3180 silencing‑induced enhancement of TSCC malignancy. In summary, the present study revealed a potential competitive endogenous RNA (ceRNA) regulatory pathway in which lncKRT16P6 modulates GATAD2A expression by binding miR‑3180, ultimately promoting tumorigenesis and metastasis in TSCC. Therefore, lncKRT16P6 may be used as a prognostic biomarker and therapeutic target for clinical intervention in TSCC.
Collapse
Affiliation(s)
- Mi Zhang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Ling Wu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Xudong Wang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| | - Jiang Chen
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian 350002, P.R. China
| |
Collapse
|
182
|
Liu Z, Jin Q, Yan T, Wo Y, Liu H, Wang Y. Exosome-mediated transduction of mechanical force regulates prostate cancer migration via microRNA. Biochem Biophys Rep 2022; 31:101299. [PMID: 35812347 PMCID: PMC9257336 DOI: 10.1016/j.bbrep.2022.101299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/29/2022] Open
Abstract
Physical cues in the extracellular microenvironment regulate cancer cell metastasis. Functional microRNA (miRNA) carried by cancer derived exosomes play a critical role in extracellular communication between cells and the extracellular microenvironment. However, little is known about the role of exosomes loaded miRNAs in the mechanical force transmission between cancer cells and extracellular microenvironment. Herein, our results suggest that stiff extracellular matrix (ECM) induced exosomes promote cancer cell migration. The ECM mechanical force regulated the exosome miRNA cargo of prostate cancer cells. Exosome miRNAs regulated by the ECM mechanical force modulated cancer cell metastasis by regulating cell motility, ECM remodeling and the interaction between cancer cells and nerves. Focal adhesion kinase mediated-ECM mechanical force regulated the intracellular miRNA expression, and F-actin mediate-ECM mechanical force regulated miRNA packaging into exosomes. The above results demonstrated that the exosome miRNA cargo promoted cancer metastasis by transmitting the ECM mechanical force. The ECM mechanical force may play multiple roles in maintaining the microenvironment of cancer metastasis through the exosome miRNA cargo. ECM mechanical force-induced exosomes regulate cancer cell migration. ECM mechanical forces regulate the cancer cell exosomes miRNA cargo. ECM mechanical forces regulated exosomes miRNAs modulate cancer metastasis by remodeling extracellular microenvironment.
Collapse
|
183
|
Palazzolo S, Canzonieri V, Rizzolio F. The history of small extracellular vesicles and their implication in cancer drug resistance. Front Oncol 2022; 12:948843. [PMID: 36091133 PMCID: PMC9451101 DOI: 10.3389/fonc.2022.948843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/01/2022] Open
Abstract
Small extracellular vesicles (EVs) in the last 20 years are demonstrated to possess promising properties as potential new drug delivery systems, biomarkers, and therapeutic targets. Moreover, EVs are described to be involved in the most important steps of tumor development and progression including drug resistance. The acquired or intrinsic capacity of cancer cells to resist chemotherapies is one of the greatest obstacles to overcome to improve the prognosis of many patients. EVs are involved in this mechanism by exporting the drugs outside the cells and transferring the drug efflux pumps and miRNAs in recipient cells, in turn inducing drug resistance. In this mini-review, the main mechanisms by which EVs are involved in drug resistance are described, giving a rapid and clear overview of the field to the readers.
Collapse
Affiliation(s)
- Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di ricovero e cura a carattere scientifico (IRCCS), Aviano, Italy
- Department of Molecular Science and Nanosystems, Ca’ Foscary University, Venice, Italy
| |
Collapse
|
184
|
Huili Y, Nie S, Zhang L, Yao A, Liu J, Wang Y, Wang L, Cao F. Cuproptosis-related lncRNA: Prediction of prognosis and subtype determination in clear cell renal cell carcinoma. Front Genet 2022; 13:958547. [PMID: 36072656 PMCID: PMC9441767 DOI: 10.3389/fgene.2022.958547] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cell carcinoma, accounting for approximately 70% of all RCC cases. Cuproptosis, a novel mechanism of cell death, may be a potential target for intervention in tumor development. Methods: Cuproptosis-related prognostic lncRNAs were identified by co-expression analysis and univariable Cox regression. Five lncRNA profiles were obtained by LASSO regression analysis, and a model with high accuracy was constructed to assess the prognosis of ccRCC patients based on these cuproptosis-related lncRNAs. Survival analysis and time-dependent ROC curves were performed for the α and β groups, and the results confirmed the high accuracy of the model in predicting the prognosis of ccRCC patients. Immunoassay, principal component analysis (PCA), and drug sensitivity analysis were also performed for different risk categories. Finally, we classified ccRCC patients into two different subtypes by consistent class clustering, and performed immune checkpoint activation, tumor microenvironment analysis, PCA, and drug sensitivity analysis for different subtypes. Results: We developed a prognostic model using five cuproptosis-associated lncRNAs, which was found to be highly accurate in predicting ccRCC patients’ prognosis. Immunotherapy may be more beneficial to the hyper-risk category and the C2 subtype. Conclusion: The results of this study confirm that five cuproptosis-associated lncRNAs can be used as potential prognostic markers for ccRCC.
Collapse
|
185
|
Feng Z, Li K, Qin K, Liang J, Shi M, Ma Y, Zhao S, Liang H, Han D, Shen B, Peng C, Chen H, Jiang L. The LINC00623/NAT10 signaling axis promotes pancreatic cancer progression by remodeling ac4C modification of mRNA. J Hematol Oncol 2022; 15:112. [PMID: 35978332 PMCID: PMC9387035 DOI: 10.1186/s13045-022-01338-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although a substantial increase in the survival of patients with other cancers has been observed in recent decades, pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest diseases. No effective screening approach exists. METHODS Differential exosomal long noncoding RNAs (lncRNAs) isolated from the serum of patients with PDAC and healthy individuals were profiled to screen for potential markers in liquid biopsies. The functions of LINC00623 in PDAC cell proliferation, migration and invasion were confirmed through in vivo and in vitro assays. RNA pulldown, RNA immunoprecipitation (RIP) and coimmunoprecipitation (Co-IP) assays and rescue experiments were performed to explore the molecular mechanisms of the LINC00623/NAT10 signaling axis in PDAC progression. RESULTS A novel lncRNA, LINC00623, was identified, and its diagnostic value was confirmed, as it could discriminate patients with PDAC from patients with benign pancreatic neoplasms and healthy individuals. Moreover, LINC00623 was shown to promote the tumorigenicity and migratory capacity of PDAC cells in vitro and in vivo. Mechanistically, LINC00623 bound to N-acetyltransferase 10 (NAT10) and blocked its ubiquitination-dependent degradation by recruiting the deubiquitinase USP39. As a key regulator of N4-acetylcytidine (ac4C) modification of mRNA, NAT10 was demonstrated to maintain the stability of oncogenic mRNAs and promote their translation efficiency through ac4C modification. CONCLUSIONS Our data revealed the role of LINC00623/NAT10 signaling axis in PDAC progression, showing that it is a potential biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Zengyu Feng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Kexian Li
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Kai Qin
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Juyong Liang
- Otolaryngology and Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yang Ma
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Shiwei Zhao
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Huaiyu Liang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, People's Republic of China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Chenghong Peng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| |
Collapse
|
186
|
Zhou M, Dong J, Huang J, Ye W, Zheng Z, Huang K, Pan Y, Cen J, Liang Y, Shu G, Ye S, Lu X, Zhang J. Chitosan-Gelatin-EGCG Nanoparticle-Meditated LncRNA TMEM44-AS1 Silencing to Activate the P53 Signaling Pathway for the Synergistic Reversal of 5-FU Resistance in Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105077. [PMID: 35717675 PMCID: PMC9353463 DOI: 10.1002/advs.202105077] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/31/2022] [Indexed: 05/16/2023]
Abstract
Chemoresistance is one of the leading causes of therapeutic failure in gastric cancer (GC) treatment. Recent studies have shown lncRNAs play pivotal roles in regulating GC chemoresistance. Nanocarriers delivery of small interfering RNAs (siRNAs) to silence cancer-related genes has become a novel approach to cancer treatment research. However, finding target genes and developing nanosystems capable of selectively delivering siRNAs for disease treatment remains a challenge. In this study, a novel lncRNA TMEM44-AS1 that is related to 5-FU resistance is identified. TMEM44-AS1 has the ability to bind to and sponge miR-2355-5p, resulting in the upregulated PPP1R13L expression and P53 pathway inhibition. Next, a new nanocarrier called chitosan-gelatin-EGCG (CGE) is developed, which has a higher gene silencing efficiency than lipo2000, to aid in the delivery of a si-TMEM44-AS1 can efficiently silence TMEM44-AS1 expression to synergistically reverse 5-FU resistance in GC, leading to a markedly enhanced 5-FU therapeutic effect in a xenograft mouse model of GC. These findings indicate that TMEM44-AS1 may estimate 5-FU therapy outcome among GC cases, and that systemic si-TMEM44-AS1 delivery combined with 5-FU therapy is significant in the treatment of patients with recurrent GC.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Catechin/analogs & derivatives
- Catechin/pharmacology
- Catechin/therapeutic use
- Cell Line, Tumor
- Chitosan/pharmacology
- Chitosan/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/physiology
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Gelatin/pharmacology
- Gelatin/therapeutic use
- Gene Expression Regulation, Neoplastic
- Gene Silencing/drug effects
- Gene Silencing/physiology
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- MicroRNAs/genetics
- Nanoparticles/therapeutic use
- RNA/genetics
- RNA/metabolism
- RNA, Antisense/genetics
- RNA, Antisense/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction/genetics
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Mi Zhou
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Jiaqi Dong
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Junqing Huang
- Guangzhou Key Laboratory of Formula‐Pattern of Traditional Chinese MedicineFormula‐Pattern Research CenterSchool of Traditional Chinese MedicineJinan UniversityGuangzhou510632P. R. China
| | - Wen Ye
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Zhousan Zheng
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Kangbo Huang
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Yihui Pan
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Junjie Cen
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Yanping Liang
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Guannan Shu
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Sheng Ye
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| | - Xuanxuan Lu
- Department of Food Science and EngineeringJinan UniversityGuangzhou510632P. R. China
| | - Jiaxing Zhang
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan road IIGuangzhou510080P. R. China
| |
Collapse
|
187
|
The Exosome Journey: From Biogenesis to Regulation and Function in Cancers. JOURNAL OF ONCOLOGY 2022; 2022:9356807. [PMID: 35898929 PMCID: PMC9313905 DOI: 10.1155/2022/9356807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/01/2022] [Accepted: 06/20/2022] [Indexed: 12/26/2022]
Abstract
Exosomes are a type of small endosomal-derived vesicles ranging from 30 to 150 nm, which can serve as functional mediators in cell-to-cell communication and various physiological and pathological processes. In recent years, exosomes have emerged as crucial mediators of intracellular communication among tumor cells, immune cells, and stromal cells, which can shuttle bioactive molecules, such as proteins, lipids, RNA, and DNA. Exosomes exhibit the high bioavailability, biological stability, targeting specificity, low toxicity, and immune characteristics, suggesting their potentials in the diagnosis and treatment of cancers. They can be applied as an effective tool in the diagnostics, therapeutics, and drug delivery in cancers. This review summarizes the regulation and functions of exosomes in various cancers to augment our understanding of exosomes, which paves the way for parallel advancements in the therapeutic approach of cancers. In this review, we also discuss the challenges and prospects for clinical application of exosome-based diagnostics and therapeutics for cancers.
Collapse
|
188
|
Xu W, Yu M, Wu Y, Jie Y, Li X, Zeng X, Yang F, Chong Y. Plasma-Derived Exosomal SncRNA as a Promising Diagnostic Biomarker for Early Detection of HBV-Related Acute-on-Chronic Liver Failure. Front Cell Infect Microbiol 2022; 12:923300. [PMID: 35873157 PMCID: PMC9301338 DOI: 10.3389/fcimb.2022.923300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives The small noncoding RNAs (sncRNAs) including microRNAs and the noncanonical sncRNAs [i.e., tRNA-derived small RNAs (tsRNAs) and rRNA-derived small RNAs (rsRNAs)] are a vital class of gene regulators in response to a variety of diseases. We focus on an sncRNA signature enriched in hepatitis B virus (HBV)-related acute-on-chronic liver failure (ACLF) to develop a plasma exosome-based noninvasive biomarker for human ACLF. Methods In this work, sncRNAs related to HBV-ACLF were identified by small RNA sequencing (RNA-seq) in plasma exosomes collected from 3 normal subjects, 4 chronic hepatitis B (CHB) patients with flare, and 6 HBV-ACLF patients in the discovery cohort. Thereafter, the differentially expressed sncRNAs were further verified in a validation cohort (n = 313) using the newly developed molecular signature incorporating different mi/ts/rsRNAs (named as MTR-RNAs) through qRT-PCR assays. Subsequently, using the least absolute shrinkage and selection operator (LASSO) logistic regression (LR) model analysis, we developed an MTR-RNA classifier for early detection of ACLF. Results The identified sncRNAs (hsa-miR-23b-3p, hsa-miR-223-3p, hsa-miR-339-5p, tsRNA-20, tsRNA-46, and rsRNA-249) were specifically differentially expressed in plasma exosomes of HBV-ACLF. The MTR-RNA signature (AUC = 0.787) containing the above sncRNAs distinguished HBV-ACLF cases among normal subjects with 71.67% specificity and 74.29% sensitivity, CHB patients with flare (AUC = 0.694, 85.71% sensitivity/59.5% specificity), and patients with CHB/cirrhosis (AUC = 0.785, 57.14% sensitivity/94.59% specificity). Notably, it revealed 100% specificity/94.80% sensitivity in detecting patients or normal people. Conclusions Our as-constructed plasma-derived exosomal sncRNA signature can serve as a reliable biomarker for ACLF detection and also be adopted to be the pre−triage biomarker for selecting cases that can gain benefits from adjuvant treatment.
Collapse
|
189
|
Thongboonkerd V, Kanlaya R. The divergent roles of exosomes in kidney diseases: Pathogenesis, diagnostics, prognostics and therapeutics. Int J Biochem Cell Biol 2022; 149:106262. [PMID: 35787447 DOI: 10.1016/j.biocel.2022.106262] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/30/2022] [Indexed: 11/16/2022]
Abstract
Exosomes are the self-packed nanoscale vesicles (nanovesicles) derived from late endosomes and released from the cells to the extracellular milieu. Exosomal biogenesis is based on endosomal pathway to form the nanovesicles surrounded by membrane originated from plasma membranes of the parental cells. During biogenesis, exosomes selectively encapsulate an array of biomolecules (proteins, nucleic acids, lipids, metabolites, etc.), thereby conveying diverse messages for cell-cell communications. Once released, these exosomal contents trigger signaling and trafficking that play roles in cell growth, development, immune responses, homeostasis, remodeling, etc. Recent advances in exosomal research have provided a wealth of useful information that enhances our knowledge on the roles for exosomes in pathogenic mechanisms of human diseases involving a wide variety of organ systems. In the kidney, exosomes play divergent roles, ranging from pathogenesis to therapeutics, based on their original sources and type of interventions. Herein, we summarize and update the current knowledge on the divergent roles of exosomes involving the pathogenesis, diagnostics, prognostics, and therapeutics in various groups of kidney diseases, including acute kidney injury, immune-mediated kidney diseases (e.g., IgA nephropathy, lupus nephritis, membranous nephropathy, focal segmental glomerulosclerosis), chronic kidney disease (caused by diabetic nephropathy and others), renal cell carcinoma, nephrolithiasis, kidney transplantation and related complications, and polycystic kidney disease. Finally, the future perspectives on research in this area are discussed.
Collapse
Affiliation(s)
- Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Rattiyaporn Kanlaya
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
190
|
Makgoo L, Mosebi S, Mbita Z. Long noncoding RNAs (lncRNAs) in HIV-mediated carcinogenesis: Role in cell homeostasis, cell survival processes and drug resistance. Noncoding RNA Res 2022; 7:184-196. [PMID: 35991514 PMCID: PMC9361211 DOI: 10.1016/j.ncrna.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/04/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
There is accruing data implicating long non-coding RNAs (lncRNAs) in the development and progression of non-communicable diseases such as cancer. These lncRNAs have been implicated in many diverse HIV-host interactions, some of which are beneficial to HIV propagation. The virus-host interactions induce the expression of HIV-regulated long non-coding RNAs, which are implicated in the carcinogenesis process, therefore, it is critical to understand the molecular mechanisms that underpin these HIV-regulated lncRNAs, especially in cancer formation. Herein, we summarize the role of HIV-regulated lncRNAs targeting cancer development-related processes including apoptosis, cell cycle, cell survival signalling, angiogenesis and drug resistance. It is unclear how lncRNAs regulate cancer development, this review also discuss recent discoveries regarding the functions of lncRNAs in cancer biology. Innovative research in this field will be beneficial for the future development of therapeutic strategies targeting long non-coding RNAs that are regulated by HIV, especially in HIV associated cancers.
Collapse
|
191
|
Ahmad E, Ali A, Nimisha, Kumar Sharma A, Apurva, Kumar A, Dar GM, Sumayya Abdul Sattar R, Verma R, Mahajan B, Singh Saluja S. Molecular markers in cancer. Clin Chim Acta 2022; 532:95-114. [DOI: https:/doi.org/10.1016/j.cca.2022.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
|
192
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
193
|
Emerging roles and potential clinical applications of long non-coding RNAs in hepatocellular carcinoma. Biomed Pharmacother 2022; 153:113327. [PMID: 35779423 DOI: 10.1016/j.biopha.2022.113327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma is one of the most common highly malignant tumors in humans, as well as the leading cause of cancer-related death worldwide. Growing evidence has indicated that lncRNAs are implicated in different molecular mechanisms, including interactions with DNA, RNA, or protein, so that to regulate the gene expression at epigenetic, transcriptional, or posttranscriptional level. Moreover, the mechanism of action of lncRNA is closely related to its subcellular localization. An increasing number of studies have certified that lncRNA plays a significant biological function in the occurrence and development of hepatocellular carcinoma, such as involving in cell proliferation, metastasis, apoptosis, ferroptosis, autophagy, and reprogramming of energy metabolism. As a result, lncRNA has great potential as a novel biomarker for diagnosis or therapeutics of hepatocellular carcinoma. In this review, we highlight the correlation between subcellular localization of lncRNA and its mechanism of action, discuss the biological roles of lncRNA and the latest research advances in hepatocellular carcinoma, and emphasize the potential of lncRNA as a therapeutic target for advanced patients of hepatocellular carcinoma.
Collapse
|
194
|
Jiang W, Li J, Cai Y, Liu W, Chen M, Xu X, Deng M, Sun J, Zhou L, Huang Y, Wu S, Cheng X. The Novel lncRNA ENST00000530525 Affects ANO1, Contributing to Blood-Brain Barrier Injury in Cultured hCMEC/D3 Cells Under OGD/R Conditions. Front Genet 2022; 13:873230. [PMID: 35754821 PMCID: PMC9213740 DOI: 10.3389/fgene.2022.873230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke (IS) is a major neurological disease with high fatality and residual disability burdens. Long noncoding RNAs (lncRNAs) have been found to play an important role in IS. However, the roles and significance of most lncRNAs in IS are still unknown. This study was performed to identify differentially expressed (DE) lncRNAs using a lncRNA microarray in whole blood samples of patients suffering from acute cerebral ischemia. Bioinformatics analyses, including GO, KEGG pathway enrichment analysis, and proximity to putative stroke risk location analysis were performed. The novel lncRNA, ENST00000530525, significantly decreased after IS. Furthermore, we evaluated lncRNA ENST00000530525 expression in cultured hCMEC/D3 cells under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions using fluorescent in situ hybridization (FISH) and quantitative real-time polymerase chain reaction (RT-qPCR) analysis. To investigate the function of lncRNA ENST00000530525, its over-expression (OE) and negative control (NC) plasmids were transfected into hCMEC/D3 cells, and cell viability was detected by a cell counting kit-8 (CCK-8) assay after OGD/R. LncRNA ENST00000530525 and ANO1 expression were investigated using RT-qPCR and immunofluorescence. For blood-brain barrier (BBB) permeability, FITC-dextran transendothelial permeability assay and tight junction (TJ) protein immunofluorescence assays were performed. There were 3352 DE lncRNAs in the blood samples of acute IS patients. The validation results were consistent with the gene chip data. The GO and KEGG results showed that these lncRNAs were mainly related to oxygen and glucose metabolism, leukocyte transendothelial migration, mitophagy and cellular senescence. Among these, lncRNA ENST00000530525 was the most highly downregulated lncRNA and it was mapped within the IS-associated gene anoctamin-1 (ANO1). We further found that lncRNA ENST00000530525 was downregulated in hCMEC/D3 cells under 4 h OGD and 20 h reoxygenation (OGD4/R20) conditions. Upregulating lncRNA ENST00000530525 by plasmid transfection decreased cell viability while increasing ANO1 expression and it contributed to BBB injury in hCMEC/D3 cells after OGD4/R20. The lncRNA ENST00000530525 might play deleterious roles in post-stroke pathogenesis. These results show that some DE lncRNAs in humans participate through characteristic roles in post-stroke pathogenesis; thus, the roles and significance of some novel lncRNAs in IS warrant further study.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jie Li
- Department of Anesthesiology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuefang Cai
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Wenchen Liu
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mei Chen
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Xu
- Department of Anatomy, Sun Yat-Sen School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Minzhen Deng
- Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingbo Sun
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China.,Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| | - Lihua Zhou
- Department of Anatomy, Sun Yat-Sen School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Yan Huang
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China.,Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| | - Shuang Wu
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Xiao Cheng
- Department of Second Institute of Clinical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China.,Department of Neurology, Guangdong Provincial Hospital of Traditional Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| |
Collapse
|
195
|
Song Q, Yu H, Han J, Qiang Lv JL, Yang H. Exosomes in urological diseases - Biological functions and clinical applications. Cancer Lett 2022; 544:215809. [PMID: 35777716 DOI: 10.1016/j.canlet.2022.215809] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/02/2022]
Abstract
Exosomes are extracellular vesicles with a variety of biological functions that exist in various biological body fluids and exert their functions through proteins, nucleic acids, lipids, and metabolites. Recent discoveries have revealed the functional and biomarker roles of miRNAs in urological diseases, including benign diseases and malignancies. Exosomes have several uses in the diagnosis, treatment, and monitoring of urological diseases, especially cancer. Proteins and nucleic acids can be used as alternative biomarkers for detecting urological diseases. Additionally, exosomes can be detected in most body fluids, thereby avoiding pathogenesis. More importantly, for urological tumors, exosomes display a higher sensitivity than circulating tumor cells and tumor-derived DNA in body fluid biopsies because of their low immunogenicity and high stability. These advantages have made it a research hotspot in recent years. In this review, we focus on the biological characteristics and functions of exosomes and summarize their advantages and the latest progress in the diagnosis and treatment of urological diseases.
Collapse
Affiliation(s)
- Qiang Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Jie Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China
| | - Jiancheng Lv Qiang Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China.
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, 210029, PR China.
| |
Collapse
|
196
|
Gao J, Zhang X, Jiang L, Li Y, Zheng Q. Tumor endothelial cell-derived extracellular vesicles contribute to tumor microenvironment remodeling. Cell Commun Signal 2022; 20:97. [PMID: 35752798 PMCID: PMC9233793 DOI: 10.1186/s12964-022-00904-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/22/2022] [Indexed: 11/12/2022] Open
Abstract
Cancer progression involves several biological steps where angiogenesis is a key tumorigenic phenomenon. Extracellular vesicles (EVs) derived from tumor cells and other cells in the tumor microenvironment (TME) help modulate and maintain favorable microenvironments for tumors. Endothelial cells (ECs) activated by cancer-derived EVs have important roles in tumor angiogenesis. Interestingly, EVs from ECs activate tumor cells, i.e. extracellular matrix (ECM) remodeling and provide more supplements for tumor cells. Thus, EV communications between cancer cells and ECs may be effective therapeutic targets for controlling cancer progression. In this review, we describe the current knowledge on EVs derived from ECs and we examine how these EVs affect TME remodeling. Video abstract
Collapse
Affiliation(s)
- Jian Gao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, China.,Science Experiment Center of China Medical University, Shenyang, 110122, China
| | - Xiaodong Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100000, China.,National Clinical Research Center for Digestive Diseases, Beijing, 100000, China
| | - Lei Jiang
- Department of General Surgery, Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yan Li
- Department of Radiotherapy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China.
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
197
|
Di Vito Nolfi M, Vecchiotti D, Flati I, Verzella D, Di Padova M, Alesse E, Capece D, Zazzeroni F. EV-Mediated Chemoresistance in the Tumor Microenvironment: Is NF-κB a Player? Front Oncol 2022; 12:933922. [PMID: 35814425 PMCID: PMC9257640 DOI: 10.3389/fonc.2022.933922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Drug resistance is a major impediment to patient survival and remains the primary cause of unsuccessful cancer therapy. Drug resistance occurs in many tumors and is frequently induced by chemotherapy which triggers a defensive response both in cancerous and cancer-associated cells that constitute the tumor microenvironment (TME). Cell to cell communication within the TME is often mediated by extracellular vesicles (EVs) which carry specific tumor-promoting factors able to activate survival pathways and immune escape mechanisms, thus sustaining tumor progression and therapy resistance. NF-κB has been recognized as a crucial player in this context. NF-κB activation is involved in EVs release and EVs, in turn, can trigger NF-κB pathway activation in specific contexts, based on secreting cytotype and their specific delivered cargo. In this review, we discuss the role of NF-κB/EVs interplay that sustain chemoresistance in the TME by focusing on the molecular mechanisms that underlie inflammation, EVs release, and acquired drug resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- *Correspondence: Francesca Zazzeroni, ; Daria Capece,
| | | |
Collapse
|
198
|
The Mystery of Exosomes in Gestational Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2169259. [PMID: 35720179 PMCID: PMC9200544 DOI: 10.1155/2022/2169259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 05/31/2022] [Indexed: 11/27/2022]
Abstract
Gestational diabetes mellitus (GDM) is one of the common pregnancy complications, which increases the risk of short-term and long-term adverse consequences in both the mother and offspring. However, the pathophysiological mechanism of GDM is still poorly understood. Inflammation, insulin resistance and oxidative stress are considered critical factors in the occurrence and development of GDM. Although the lifestyle intervention and insulin are the primary treatment, adverse pregnancy outcomes still cannot be ignored. Exosomes have a specific function of carrying biological information, which can transmit information to target cells and play an essential role in intercellular communication. Their possible roles in normal pregnancy and GDM have been widely concerned. The possibility of exosomal cargos as biomarkers of GDM is proposed. This paper reviews the literature in recent years and discusses the role of exosomes in GDM and their possible mechanisms to provide some reference for the prediction, prevention, and treatment of GDM and improve the outcome of pregnancy.
Collapse
|
199
|
Badowski C, He B, Garmire LX. Blood-derived lncRNAs as biomarkers for cancer diagnosis: the Good, the Bad and the Beauty. NPJ Precis Oncol 2022; 6:40. [PMID: 35729321 PMCID: PMC9213432 DOI: 10.1038/s41698-022-00283-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/13/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer ranks as one of the deadliest diseases worldwide. The high mortality rate associated with cancer is partially due to the lack of reliable early detection methods and/or inaccurate diagnostic tools such as certain protein biomarkers. Cell-free nucleic acids (cfNA) such as circulating long noncoding RNAs (lncRNAs) have been proposed as a new class of potential biomarkers for cancer diagnosis. The reported correlation between the presence of tumors and abnormal levels of lncRNAs in the blood of cancer patients has notably triggered a worldwide interest among clinicians and oncologists who have been actively investigating their potentials as reliable cancer biomarkers. In this report, we review the progress achieved ("the Good") and challenges encountered ("the Bad") in the development of circulating lncRNAs as potential biomarkers for early cancer diagnosis. We report and discuss the diagnostic performance of more than 50 different circulating lncRNAs and emphasize their numerous potential clinical applications ("the Beauty") including therapeutic targets and agents, on top of diagnostic and prognostic capabilities. This review also summarizes the best methods of investigation and provides useful guidelines for clinicians and scientists who desire conducting their own clinical studies on circulating lncRNAs in cancer patients via RT-qPCR or Next Generation Sequencing (NGS).
Collapse
Affiliation(s)
- Cedric Badowski
- University of Hawaii Cancer Center, Epidemiology, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| | - Bing He
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Lana X Garmire
- University of Hawaii Cancer Center, Epidemiology, 701 Ilalo Street, Honolulu, HI, 96813, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
200
|
Ma T, Wu FH, Wu HX, Fa Q, Chen Y. Long Non-Coding RNA MCM3AP-AS1: A Crucial Role in Human Malignancies. Pathol Oncol Res 2022; 28:1610194. [PMID: 35783356 PMCID: PMC9243217 DOI: 10.3389/pore.2022.1610194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/31/2022] [Indexed: 12/09/2022]
Abstract
The incidence of cancer continues to grow and is one of the leading causes of death in the world. Long noncoding RNAs (LncRNAs) is a group of RNA transcripts greater than 200 nucleotides in length, and although it cannot encode proteins, it can regulate different biological functions by controlling gene expression, transcription factors, etc. LncRNA micro-chromosome maintenance protein 3-associated protein antisense RNA 1 (MCM3AP-AS1) is involved in RNA processing and cell cycle-related functions, and MCM3AP-AS1 is dysregulated in expression in various types of cancers. This biomarker is involved in many processes related to carcinogens, such as cell proliferation, apoptosis, cell cycle, and migration. In this review, we summarize the roles of MCM3AP-AS1 in different human cancers and its biological functions with a view to providing ideas for future research.
Collapse
Affiliation(s)
- Tao Ma
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fa-Hong Wu
- Department of General Surgery Hepatic-Biliary-Pancreatic Institute, Lanzhou University Second Hospital, Lanzhou, China
| | - Hong-Xia Wu
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiong Fa
- Department of Nuclear Medicine, The 940th Hospital of the People’s Liberation Army Joint Service Support Force, Lanzhou, China
| | - Yan Chen
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Yan Chen,
| |
Collapse
|