1
|
Ge Z, Zhang P, Xia Y, Lu C, Sun C, Wen Z, Chen W, Li Y, Lin S, Wu Y, Li X, Zhou H, Wang W, Chen S, Ji L, Lai Y. A three-miRNA panel in serum: Serving as a novel diagnostic method for nasopharyngeal carcinoma. Int J Biol Markers 2025:3936155251329041. [PMID: 40232237 DOI: 10.1177/03936155251329041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
BackgroundNasopharyngeal carcinoma has unique epidemiological characteristics. Screening for this currently lacks a highly efficient, non-invasive, and inexpensive method. Serum microRNA (miRNA), which is stable and commonly present, has the potential to serve as a novel marker for nasopharyngeal carcinoma diagnosis.ObjectivesThis study aims to find a highly efficient, non-invasive, and inexpensive biomarker for nasopharyngeal carcinoma diagnosis.MethodsThis study, involving 52 patients with nasopharyngeal carcinoma and 56 healthy controls, was conducted in two phases to identify miRNAs in the serum suitable for nasopharyngeal carcinoma diagnosis using quantitative reverse transcription polymerase chain reaction. Stepwise logistic regression analysis was then used to identify a miRNA panel with high diagnostic efficiency. Additionally, we used bioinformatic analysis to explore the potential biological functions of the crucial miRNAs.ResultsA three-miRNA panel (miR-148b-3p, miR-10b-5p, and miR-18a-5p) has a high diagnostic value for nasopharyngeal carcinoma (area under the curve = 0.872; 95% confidence interval: 0.793-0.928; sensitivity = 78.57%; specificity = 86.54%). Through bioinformatics analysis we found that CC2D2B, PCDH9, and FOXP1 may be potential target genes of these three miRNAs.ConclusionThis three-miRNA panel (miR-148b-3p, miR-10b-5p, and miR-18a-5p) represents a highly efficient, non-invasive, and inexpensive biomarker for diagnosing nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Zhenjian Ge
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- PKU-Shenzhen Clinical Institute of Shantou University Medical College, Guangdong, China
| | - Pengwu Zhang
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
| | - Yong Xia
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Guangdong, China
| | - Chong Lu
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Chen Sun
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Zhenyu Wen
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- PKU-Shenzhen Clinical Institute of Shantou University Medical College, Guangdong, China
| | - Wenkang Chen
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- PKU-Shenzhen Clinical Institute of Shantou University Medical College, Guangdong, China
| | - Yingqi Li
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- Shenzhen University Health Science Center, Guangdong, China
| | - Shengjie Lin
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- Shenzhen University Health Science Center, Guangdong, China
| | - Yutong Wu
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- PKU-Shenzhen Clinical Institute of Shantou University Medical College, Guangdong, China
| | - Xutai Li
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Huimei Zhou
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Wuping Wang
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- Shenzhen University Health Science Center, Guangdong, China
| | - Siwei Chen
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
- Shenzhen University Health Science Center, Guangdong, China
| | - Ling Ji
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Guangdong, China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Guangdong, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Guangdong, China
| |
Collapse
|
2
|
Mangani S, Piperigkou Z, Koletsis NE, Ioannou P, Karamanos NK. Estrogen receptors and extracellular matrix: the critical interplay in cancer development and progression. FEBS J 2025; 292:1558-1572. [PMID: 39285617 PMCID: PMC11970714 DOI: 10.1111/febs.17270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/26/2024] [Accepted: 09/02/2024] [Indexed: 04/05/2025]
Abstract
Cancer remains a significant global health concern. Breast cancer is a multifaceted and prevalent disease influenced by several factors, among which estrogen receptors (ERs) and the extracellular matrix (ECM) play pivotal roles. ERs, encompassing ERα and ERβ, exert significant diversity on tumor behavior, cell signaling, invasion, and metastatic potential, thus guiding breast cancer prognosis. Understanding the multifunctional connections between ERs and ECM that mediate the dynamics of tumor microenvironment is vital for unraveling the complexity of breast cancer pathobiology and identifying novel therapeutic targets. This critical review delves into the intricate nature of ERs, emphasizing their structural isoforms and the consequential impact on breast cancer outcomes. A detailed examination of ER-mediated cell signaling pathways reveals how differential expression of ERα and ERβ isoforms influence breast cancer cell behavior. The functional ERs-matrix interactions emerge as a pivotal factor in modulating epigenetic mechanisms of breast cancer cells, orchestrating changes in cellular phenotype and expression patterns of matrix modulators. Specifically, ERα isoforms are shown to regulate ECM signaling cascades, while the effects of ECM components on ERα activity highlight a bidirectional regulatory axis. The diversity of ERβ isoforms is also highlighted, illustrating their distinct contribution to ECM-mediated cellular responses. This review underscores the complex interplay between ERα/β isoforms and the ECM, shedding light onto the potential therapeutic strategies targeting these interactions to improve breast cancer management.
Collapse
Affiliation(s)
- Sylvia Mangani
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Nikolaos E. Koletsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Paraskevi Ioannou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of ChemistryUniversity of PatrasGreece
| |
Collapse
|
3
|
Wang Y, Wang S, He H, Bai Y, Liu Z, Sabihi SS. Mechanisms of apoptosis-related non-coding RNAs in ovarian cancer: a narrative review. Apoptosis 2025; 30:553-578. [PMID: 39833637 DOI: 10.1007/s10495-024-02074-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Ovarian cancer remains a major challenge in oncology due to its complex biology and late-stage diagnosis. Recent advances in molecular biology have highlighted the crucial role of non-coding RNAs (ncRNAs) in regulating apoptosis and cancer progression. NcRNAs, including microRNAs, long non-coding RNAs, and circular RNAs, have emerged as significant players in the molecular networks governing ovarian cancer. Despite these insights, the precise mechanisms by which ncRNAs influence ovarian cancer pathology are not fully understood. This complexity, combined with the heterogeneity of the disease and the development of treatment resistance, poses substantial obstacles to effective therapeutic development. Additionally, the lack of reliable early detection methods further complicates treatment strategies. This manuscript reviews the current state of research on ncRNAs in ovarian cancer, discusses the challenges in translating these findings into clinical applications, and outlines potential future directions. Emphasis is placed on the need for integrated approaches to unravel the intricate roles of ncRNAs, improve early detection, and develop personalized treatment strategies to address the diverse and evolving nature of ovarian cancer. While these findings provide valuable insights, it is crucial to recognize that many results are based on preclinical studies and require further validation to establish their clinical applicability.
Collapse
Affiliation(s)
- Yue Wang
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Shirui Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710038, China
| | - Haiyan He
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Yingying Bai
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Zhuo Liu
- Department of Obstetrics and Gynecology, Xi'an International Medical Center Hospital, Xi'an, 710038, China
| | - Sima-Sadat Sabihi
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
4
|
Ge Z, Lin S, Lu C, Xia Y, Li R, Li X, Sun C, Wen Z, Chen W, Li Y, Li M, Lin Y, Dong J, Tao L, Ji L, Lai Y. Panel containing three serum microRNAs: a promising biomarker for early detection of bladder cancer. Front Oncol 2024; 14:1470457. [PMID: 39777350 PMCID: PMC11703750 DOI: 10.3389/fonc.2024.1470457] [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: 07/25/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Background Bladder cancer (BC) is a common tumor worldwide. Screening for BC currently lacks a highly efficient, non-invasive, and inexpensive method. Serum microRNA (miRNA), which is stable and commonly present, has the potential to serve as a novel marker for BC diagnosis. Materials & methods Based on a study involving 112 BC patients and 112 healthy subjects, we conducted this research in three phases to identify applicable microRNAs (miRNAs) in serum for BC diagnosis using quantitative reverse transcription polymerase chain reaction (qRT-PCR). A panel with optimal diagnostic value was developed. Additionally, we used bioinformatic analysis to explore the potential biological functions of the crucial miRNAs. Results The diagnostic panel consisted of miR-212-3p, miR-30c-5p, and miR-206, with an area under the curve (AUC) of 0.838, sensitivity of 83.33%, and specificity of 73.81%. Furthermore, ATF3, GJA1, JPH2, MVB12B, RUNX1T1, SLC8A1, SPATA6, and TPM3 may be potential target genes of these three miRNAs. Conclusion We developed a three-miRNA panel that could serve as a highly efficient and inexpensive biomarker for BC diagnosis and screening.
Collapse
Affiliation(s)
- Zhenjian Ge
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Shengjie Lin
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Chong Lu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Yong Xia
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rongkang Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Xinji Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Chen Sun
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- The Fifth Clinical Medical College of Anhui Medical University, Hefei, China
| | - Zhenyu Wen
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Wenkang Chen
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shantou University Medical College, Shantou, China
| | - Yingqi Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
- Shenzhen University Health Science Center, Shenzhen, China
| | - Mingyang Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yu Lin
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Dong
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lingzhi Tao
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
| | - Ling Ji
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Urology, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, China
| |
Collapse
|
5
|
Wang Z, Wu Y, Wang Z, Tian Z, Liu Y, Liu S. In situ noninvasive monitoring of cell secretions based on MOFs/AAO hybrid membrane induced asymmetric ion transport. Biosens Bioelectron 2024; 266:116735. [PMID: 39241337 DOI: 10.1016/j.bios.2024.116735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Nanofluidic hybrid membranes display distinct ionic current rectification (ICR) properties and provide high surface area for immobilizing probes on the outer surface, exhibiting great potential in detection of biomolecules. Herein, we fabricated MOFs/AAO hybrid membrane with aptamers functionalized on the outer surface for in situ detection of living cells released secretions. TNF-α (a small molecular protein secreted by macrophages) was used as a model. After TNF-α was specifically captured by aptamers on the membrane surface, the asymmetry of surface charge on the hybrid membrane was amplified, the ICR was increased from 3.89 to 18.85. According to the ICR change, TNF-α was sensitively measured with a detection limit of ∼0.49 pM, which was significantly lower than other reported methods. When the hybrid membrane was clamped in the middle of self-made device, PET membrane incubated macrophages was rolled up and inserted into the chamber to mimic cellular microenvironment. Macrophages released TNF-α could be real time monitored with ionic current, macrophages and normal cells could be effectively distinguished according to the released TNF-α level. Thus, we proposed a nanofluidic platform for accurately measuring cell secretions in an engineered cellular microenvironment with a direct manner, without the need for labels or amplification steps.
Collapse
Affiliation(s)
- Zhaohan Wang
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Zhi Wang
- Wuxi Institute of Inspection, Testing and Certification, Wuxi 214125, China
| | - Zhaoyan Tian
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Yu Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
6
|
Nappi F, Nassif A, Schoell T. External Scaffold for Strengthening the Pulmonary Autograft in the Ross Procedure. Biomimetics (Basel) 2024; 9:674. [PMID: 39590246 PMCID: PMC11591583 DOI: 10.3390/biomimetics9110674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Despite offering several potential benefits over standard prosthetic aortic valve replacement, the use of the pulmonary autograft has been limited to date due to concerns over the risk of pulmonary autograft expansion and the need for reintervention. Several techniques using materials with biomimetic potential have been developed to reduce this complication. The incidence, risk factors, and pathophysiology of pulmonary autograft dilatation are discussed in this article. This seminar will provide an overview of the techniques of external pulmonary autograft support and their advantages and limitations. It also considers future directions for further investigation and future clinical applications of external pulmonary autograft support. Dilatation of the autograft is more likely to occur in patients with aortic regurgitation and a dilated aortic annulus. External scaffolding may prevent autograft stretching and expansion in these specific cases. However, from a biomimetic point of view, any permanent scaffold potentially restricts the movement of the autograft root. This reduces some of the benefits associated with the use of autologous tissue, which is the priority of the Ross procedure. To address this issue, several bioresorbable matrices could be used to support the root during its initial adaptive phase. Control of blood pressure with aggressive therapy is the first line to avoid this problem in the first year after pulmonary autograft implantation, together with support of the annular and sinotubular junction in some selected cases. This is the best way to maintain stable autograft root dimensions while preserving root dynamics. However, to determine the efficacy of this combined external support and best medical management, it is important to perform regular imaging and clinical follow-up.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.N.); (T.S.)
| | | | | |
Collapse
|
7
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
8
|
Kaleem M, Thool M, Dumore NG, Abdulrahman AO, Ahmad W, Almostadi A, Alhashmi MH, Kamal MA, Tabrez S. Management of triple-negative breast cancer by natural compounds through different mechanistic pathways. Front Genet 2024; 15:1440430. [PMID: 39130753 PMCID: PMC11310065 DOI: 10.3389/fgene.2024.1440430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most severe form of breast cancer, characterized by the loss of estrogen, progesterone, and human epidermal growth factor receptors. It is caused by various genetic and epigenetic factors, resulting in poor prognosis. Epigenetic changes, such as DNA methylation and histone modification, are the leading mechanisms responsible for TNBC progression and metastasis. This review comprehensively covers the various subtypes of TNBC and their epigenetic causes. In addition, the genetic association of TNBC with all significant genes and signaling pathways linked to the progression of this form of cancer has been enlisted. Furthermore, the possible uses of natural compounds through different mechanistic pathways have also been discussed in detail for the successful management of TNBC.
Collapse
Affiliation(s)
- Mohammed Kaleem
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra, India
| | - Mandar Thool
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Nagpur, Maharashtra, India
| | - Nitin G. Dumore
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra, India
| | | | - Wasim Ahmad
- Department of KuliyateTib, National Institute of Unani Medicine, Bengaluru, India
| | - Amal Almostadi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Hassan Alhashmi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for High Altitude Medicine, Institutes for Systems Genetics, West China School of Nursing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Birulia, Bangladesh
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Enzymoics, Hebersham, NSW, Australia; Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
Kang D, Kim T, Choi GE, Park A, Yoon J, Yu J, Suh N. miR-29a-3p orchestrates key signaling pathways for enhanced migration of human mesenchymal stem cells. Cell Commun Signal 2024; 22:365. [PMID: 39020373 PMCID: PMC11256664 DOI: 10.1186/s12964-024-01737-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The homing of human mesenchymal stem cells (hMSCs) is crucial for their therapeutic efficacy and is characterized by the orchestrated regulation of multiple signaling modules. However, the principal upstream regulators that synchronize these signaling pathways and their mechanisms during cellular migration remain largely unexplored. METHODS miR-29a-3p was exogenously expressed in either wild-type or DiGeorge syndrome critical region 8 (DGCR8) knockdown hMSCs. Multiple pathway components were analyzed using Western blotting, immunohistochemistry, and real-time quantitative PCR. hMSC migration was assessed both in vitro and in vivo through wound healing, Transwell, contraction, and in vivo migration assays. Extensive bioinformatic analyses using gene set enrichment analysis and Ingenuity pathway analysis identified enriched pathways, upstream regulators, and downstream targets. RESULTS The global depletion of microRNAs (miRNAs) due to DGCR8 gene silencing, a critical component of miRNA biogenesis, significantly impaired hMSC migration. The bioinformatics analysis identified miR-29a-3p as a pivotal upstream regulator. Its overexpression in DGCR8-knockdown hMSCs markedly improved their migration capabilities. Our data demonstrate that miR-29a-3p enhances cell migration by directly inhibiting two key phosphatases: protein tyrosine phosphatase receptor type kappa (PTPRK) and phosphatase and tensin homolog (PTEN). The ectopic expression of miR-29a-3p stabilized the polarization of the Golgi apparatus and actin cytoskeleton during wound healing. It also altered actomyosin contractility and cellular traction forces by changing the distribution and phosphorylation of myosin light chain 2. Additionally, it regulated focal adhesions by modulating the levels of PTPRK and paxillin. In immunocompromised mice, the migration of hMSCs overexpressing miR-29a-3p toward a chemoattractant significantly increased. CONCLUSIONS Our findings identify miR-29a-3p as a key upstream regulator that governs hMSC migration. Specifically, it was found to modulate principal signaling pathways, including polarization, actin cytoskeleton, contractility, and adhesion, both in vitro and in vivo, thereby reinforcing migration regulatory circuits.
Collapse
Affiliation(s)
- Dayeon Kang
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Taehwan Kim
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Ga-Eun Choi
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Arum Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jin Yoon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinho Yu
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Nayoung Suh
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
10
|
Qiu H, Fu Y, Guo Z, Zhang X, Wang X, Wu H. Dysregulated microRNAs and long non-coding RNAs associated with extracellular matrix stiffness. Exp Cell Res 2024; 437:114014. [PMID: 38547959 DOI: 10.1016/j.yexcr.2024.114014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Extracellular matrix (ECM) stiffness regulates development and homeostasis in vivo and affects both physiological and pathological processes. A variety of studies have demonstrated that mRNAs, such as Piezo1, integrin β1, and Yes-associated protein (YAP)/tafazzin (TAZ), can sense the mechanical signals induced by ECM stiffness and transmit them from the extracellular space into the cytoplasm. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), have been reported to play important roles in various cellular processes. Therefore, the interactions between ncRNAs and ECM stiffness, as well as the underlying molecular mechanisms, have become intriguing. In this review, we summarize recent findings on miRNAs and lncRNAs that interact with ECM stiffness. Several miRNAs and lncRNAs are involved in the progression of liver cancer, breast cancer, osteosarcoma, and cardiovascular diseases under the regulation of ECM stiffness. Through these ncRNAs, cellular behaviors including cell differentiation, proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) are affected by ECM stiffness. We also integrate the ncRNA signaling pathways associated with ECM stiffness, in which typical signaling pathways like integrin β1/TGFβ1, phosphatidylinositol-3 kinase (PI3K)/AKT, and EMT are involved. Although our understanding of the relationships between ncRNAs and ECM stiffness is still limited, further investigations may provide new insights for disease treatment. ECM-associated ncRNAs may serve as disease biomarkers or be targeted by drugs.
Collapse
Affiliation(s)
- Huimin Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Yangpu, 200093, Shanghai, China; Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Yi Fu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Zhinan Guo
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China; School of Sports and Health, Shanghai University of Sport, Yangpu, 200438, Shanghai, China.
| | - Xinjia Zhang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Xinyue Wang
- School of Medical Instruments, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| | - Hailong Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Pudong, 201318, Shanghai, China.
| |
Collapse
|
11
|
Monteiro MM, Gomes CC, Cruz MC, Horliana ACRT, Hamassaki DE, Lima CR, Santos MF. High glucose impairs human periodontal ligament cells migration through lowered microRNAs 221 and 222. J Periodontal Res 2024; 59:336-345. [PMID: 38041212 DOI: 10.1111/jre.13217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 11/02/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE To investigate the effects of miR-221 and miR-222 and high glucose on human periodontal ligament (PL) cells morphology, cytoskeleton, adhesion, and migration. BACKGROUND Chronic hyperglycemia is common in uncontrolled diabetes mellitus (DM) and plays a central role in long-term DM complications, such as impaired periodontal healing. We have previously shown that high glucose increases apoptosis of human PL cells by inhibiting miR-221 and miR-222 and consequently augmenting their target caspase-3. However, other effects of miR-221/222 downregulation on PL cells are still unknown. METHODS Cells from young humans' premolar teeth were cultured for 7 days under 5 or 30 mM glucose. Directional and spontaneous migration on fibronectin were studied using transwell and time-lapse assays, respectively. F-actin staining was employed to study cell morphology and the actin cytoskeleton. MiR-221 and miR-222 were inhibited using antagomiRs, and their expressions were evaluated by real-time RT-PCR. RESULTS High glucose inhibited PL cells early adhesion, spreading, and migration on fibronectin. Cells exposed to high glucose showed reduced polarization, velocity, and directionality. They formed several simultaneous unstable and short-lived protrusions, suggesting impairment of adhesion maturation. MiR-221 and miR-222 inhibition also reduced migration, decreasing cell directionality but not significantly cell velocity. After miR-221 and miR-222 downregulation cells showed morphological resemblance with cells exposed to high glucose. CONCLUSION High glucose impairs human PL cells migration potentially through a mechanism involving reduction of microRNA-221 and microRNA-222 expression. These effects may contribute to the impairment of periodontal healing, especially after surgery and during guided regeneration therapies.
Collapse
Affiliation(s)
- Mariana Marin Monteiro
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Cibele Crastequini Gomes
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mario Costa Cruz
- Center of Facilities for Research Support (CEFAP-USP), Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | | | - Dânia Emi Hamassaki
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Cilene Rebouças Lima
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Marinilce F Santos
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Ma L, Li C, Yin H, Huang J, Yu S, Zhao J, Tang Y, Yu M, Lin J, Ding L, Cui Q. The Mechanism of DNA Methylation and miRNA in Breast Cancer. Int J Mol Sci 2023; 24:9360. [PMID: 37298314 PMCID: PMC10253858 DOI: 10.3390/ijms24119360] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/17/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Breast cancer is the most prevalent cancer in the world. Currently, the main treatments for breast cancer are radiotherapy, chemotherapy, targeted therapy and surgery. The treatment measures for breast cancer depend on the molecular subtype. Thus, the exploration of the underlying molecular mechanisms and therapeutic targets for breast cancer remains a hotspot in research. In breast cancer, a high level of expression of DNMTs is highly correlated with poor prognosis, that is, the abnormal methylation of tumor suppressor genes usually promotes tumorigenesis and progression. MiRNAs, as non-coding RNAs, have been identified to play key roles in breast cancer. The aberrant methylation of miRNAs could lead to drug resistance during the aforementioned treatment. Therefore, the regulation of miRNA methylation might serve as a therapeutic target in breast cancer. In this paper, we reviewed studies on the regulatory mechanisms of miRNA and DNA methylation in breast cancer from the last decade, focusing on the promoter region of tumor suppressor miRNAs methylated by DNMTs and the highly expressed oncogenic miRNAs inhibited by DNMTs or activating TETs.
Collapse
Affiliation(s)
- Lingyuan Ma
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Chenyu Li
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Hanlin Yin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Jiashu Huang
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Shenghao Yu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Jin Zhao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Yongxu Tang
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Min Yu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Jie Lin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Lei Ding
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| | - Qinghua Cui
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.M.); (C.L.); (H.Y.); (J.H.); (S.Y.); (J.Z.); (Y.T.); (M.Y.); (J.L.)
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, Kunming 650214, China
| |
Collapse
|
13
|
Piperigkou Z, Karamanos NK. Evaluating the Effects of MicroRNAs on Proteoglycans and Matrix Constituents' Expression and Functional Properties. Methods Mol Biol 2023; 2619:257-271. [PMID: 36662476 DOI: 10.1007/978-1-0716-2946-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Circulating microRNAs (miRNAs) possess significant roles in normal homeostasis and disease conditions, including cardiovascular diseases, fibrosis, inflammatory response, and cancer. Secreted miRNAs, via the membrane vesicles, actively communicate with extracellular matrix (ECM) components to affect cell-cell and cell-matrix interactions, thereby affecting matrix remodeling and metabolic pathways in the recipient cells. Matrix macromolecules regulate the expression of miRNAs, and in turn miRNAs have been identified as emerging mediators of matrix constituents, serving as appealing biomarkers for many pathophysiological processes. Therefore, the expression profile of certain miRNAs highlights the importance of their targeting in several aspects of human pathologies. In this chapter, we report molecular biology protocols to determine the effects of selected miRNAs on the expression and activity of matrix biomolecules.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece.
| |
Collapse
|
14
|
Cao G, Miao H, Wang YN, Chen DQ, Wu XQ, Chen L, Guo Y, Zou L, Vaziri ND, Li P, Zhao YY. Intrarenal 1-methoxypyrene, an aryl hydrocarbon receptor agonist, mediates progressive tubulointerstitial fibrosis in mice. Acta Pharmacol Sin 2022; 43:2929-2945. [PMID: 35577910 PMCID: PMC9622813 DOI: 10.1038/s41401-022-00914-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/17/2022] [Indexed: 12/29/2022]
Abstract
Recent studies have shown that endogenous metabolites act via aryl hydrocarbon receptor (AhR) signalling pathway in tubulointerstitial fibrosis (TIF) pathogenesis. However, the mechanisms underlying endogenous metabolite-mediated AhR activation are poorly characterised. In this study, we conducted untargeted metabolomics analysis to identify the significantly altered intrarenal metabolites in a mouse model of unilateral ureteral obstruction (UUO). We found that the levels of the metabolite 1-methoxypyrene (MP) and the mRNA expression of AhR and its target genes CYP1A1, CYP1A2, CYP1B1 and COX-2 were progressively increased in the obstructed kidney at Weeks 1, 2 and 3. Furthermore, these changes were positively correlated with progressive TIF in UUO mice. In NRK-52E, RAW 264.7 and NRK-49F cells, MP dose-dependently upregulated the mRNA expression of AhR and its four target genes and the protein expression of nuclear AhR, accompanied by the upregulated protein expression of collagen I, α-SMA and fibronectin, as well as downregulated E-cadherin expression. Consistently, oral administration of MP in mice progressively enhanced AhR activity and upregulated profibrotic protein expression in the kidneys; these effects were partially inhibited by AhR knockdown in MP-treated mice and cell lines. In addition, we screened and identified erythro-guaiacylglycerol-β-ferulic acid ether (GFA), which was isolated from Semen plantaginis, as a new AhR antagonist. GFA significantly attenuated TIF in MP-treated NRK-52E cells and mice by partially antagonising AhR activity. Our results suggest that MP activates AhR signalling, thus mediating TIF through epithelial-mesenchymal transition and macrophage-myofibroblast transition. MP is a crucial metabolite that contributes to TIF via AhR signalling pathway.
Collapse
Affiliation(s)
- Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China.
| | - Hua Miao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China
| | - Yan-Ni Wang
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China
| | - Dan-Qian Chen
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, Department of Nephrology, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Beijing, 100029, China
| | - Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China
| | - Lin Chen
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, 1700 Lomas Blvd NE, Albuquerque, NM, 87131, USA
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, No. 2025 Chengluo Avenue, Chengdu, 610106, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA, 92897, USA
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, Department of Nephrology, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Beijing, 100029, China.
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, 310053, China.
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
15
|
A Cell Component-Related Prognostic Signature for Head and Neck Squamous Cell Carcinoma Based on the Tumor Microenvironment. Int J Genomics 2022; 2022:6022869. [PMID: 35795712 PMCID: PMC9252828 DOI: 10.1155/2022/6022869] [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: 05/06/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous disease with a high mortality rate. The tumor microenvironment (TME) is composed of numerous noncancerous cells that contribute to tumorigenesis and prediction of therapeutic effects. In this study, we aimed to develop a cell component-related prognostic model based on TME. We screened cell component enrichments from samples in The Cancer Genome Atlas (TCGA) HNSCC cohort using the xCell algorithm. Univariate Cox and multivariate Cox regression analyses were performed to establish an optimal independent risk model. The prognostic value of the model was further validated using Gene Expression Omnibus datasets. We found that patients in the low-risk group had a better outcome and activated immunity and may benefit more from the immune checkpoint inhibitor therapy. We also explored microRNAs (miRNAs) that may regulate these identified cell components, and 11 miRNA expression levels influenced the overall survival time. Moreover, their target mRNAs were differentially expressed in TCGA cohort and enriched in pathways of cell cycle pathways, extracellular matrix receptor interaction, human papillomavirus infection, and cancer. In summary, our cell component-related signature was a promising prognostic biomarker that provides new insights into the predictive value of nontumor components in the TME.
Collapse
|
16
|
Scuruchi M, D'Ascola A, Avenoso A, Zappone A, Mandraffino G, Campo S, Campo GM. miR9 inhibits 6-mer HA-induced cytokine production and apoptosis in human chondrocytes by reducing NF-kB activation. Arch Biochem Biophys 2022; 718:109139. [PMID: 35114139 DOI: 10.1016/j.abb.2022.109139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 01/21/2022] [Accepted: 01/29/2022] [Indexed: 02/07/2023]
Abstract
The present study aimed to investigate the expression of miR9 and its correlation with cytokines, proteolytic enzymes and apoptosis in an experimental model of 6-mer HA induced inflammation in human chondrocytes. Human articular chondrocytes, transfected with a miR-9 mimic and miR-9 inhibitor, were stimulated with 6-mer HA in presence/absence of a specific NF-kB inhibitor. 6-mer HA induced a significant increase of TLR-4, CD44, IL-8, IL-18, MMP-9, ADAMTS-5, BAX and BCL-2 mRNAs expression and the related proteins, as well as NF-kB activation, associated with a significant up regulation of miR-9. In chondrocytes transfected with the miR-9 mimic before 6-mer HA treatment we found a decrease of such inflammatory cytokines, metalloproteases and pro-apoptotic molecules, while we found them increased in chondrocytes transfected with the miR9 inhibitor before 6-mer HA stimulation. The activities of TLR-4 and CD44, up regulated by 6-mer HA, were not modified by miR9 mimic/inhibitor, while the NF-kB activation was significantly affected. We suggested that the up regulation of miR9, induced by 6-mer HA, could be a cellular attempt to limit cell damage during inflammation.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Policlinico Universitario, 98125, Messina, Italy.
| | - Angela D'Ascola
- Department of Clinical and Experimental Medicine, University of Messina, Policlinico Universitario, 98125, Messina, Italy
| | - Angela Avenoso
- Department of Biomedical and Dental Sciences and Morphofunctional Images, Policlinico Universitario, University of Messina, 98125, Messina, Italy
| | - Annie Zappone
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, Policlinico Universitario, 98125, Messina, Italy
| | - Salvatore Campo
- Department of Biomedical and Dental Sciences and Morphofunctional Images, Policlinico Universitario, University of Messina, 98125, Messina, Italy
| | - Giuseppe M Campo
- Department of Clinical and Experimental Medicine, University of Messina, Policlinico Universitario, 98125, Messina, Italy
| |
Collapse
|
17
|
Piperigkou Z, Tzaferi K, Makrokanis G, Cheli K, Karamanos NK. The microRNA-cell surface proteoglycan axis in cancer progression. Am J Physiol Cell Physiol 2022; 322:C825-C832. [PMID: 35294845 DOI: 10.1152/ajpcell.00041.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proteoglycans consist one of the major extracellular matrix class of biomolecules that demonstrate nodal roles in cancer progression. Μodern diagnostic and therapeutic approaches include proteoglycan detection and pharmacological targeting in various cancers. Proteoglycans orchestrate critical signaling pathways for cancer development and progression through dynamic interactions with matrix components. It is well established that the epigenetic signatures of cancer cells play critical role in guiding their functional properties and metastatic potential. Secreted microRNAs (miRNAs) reside in a complex network with matrix proteoglycans, thus affecting cell-cell and cell-matrix communication. This mini-review aims to highlight current knowledge on the proteoglycan-mediated signaling cascades that regulate miRNA biogenesis in cancer. Moreover, the miRNA-mediated proteoglycan regulation during cancer progression and mechanistic aspects on the way that proteoglycans affect miRNA expression are presented. Recent advances on the role of cell surface proteoglycans in exosome biogenesis and miRNA packaging and expression are also discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Kyriaki Tzaferi
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - George Makrokanis
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konsatntina Cheli
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
18
|
Li R, Lu C, Yang W, Zhou Y, Zhong J, Chen X, Li X, Huang G, Peng X, Liu K, Zhang C, Hu H, Lai Y. A panel of three serum microRNA can be used as potential diagnostic biomarkers for nasopharyngeal carcinoma. J Clin Lab Anal 2022; 36:e24194. [PMID: 35028969 PMCID: PMC8842135 DOI: 10.1002/jcla.24194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma is cancer with unique epidemiological characteristics, showing obvious ethnicity, gender, and geographical prevalence. More and more evidence shows that microRNAs are stable in serum and are specific to different tumor types. Therefore, miRNA is a new non-invasive biomarker for cancer detection. METHODS The experiment is divided into three stages, namely, the screening stage, the training stage, and the verification stage. We took 54 patients with nasopharyngeal carcinoma and 108 healthy controls as the research objects. We use the receiver-operating characteristic (ROC) curve and area under the ROC curve (AUC) to evaluate the diagnostic value of miRNA. Finally, a three-miRNA panel with high diagnostic efficiency was constructed. In addition, we conducted biological information analysis of these miRNAs to explore their functions. RESULTS In NPC patients, the expression of five serum miRNAs (miR-29c-3p, miR-143-5p, miR-150-5p, miR-145-3p, and miR-205-5p) is significantly dysregulated. Among them, the diagnostic value of these three miRNAs (miR-29c-3p, AUC = 0.702; miR-143-5p, AUC = 0.733; and miR-205-5p, AUC = 787) is more prominent. The diagnostic panel constructed by them has a higher diagnostic value (AUC = 0.902). Through the analysis of the TCGA data set, the target gene of the three-miRNA panel may be KLF7, NRG1, SH3BGRL2, and SYNPO2. CONCLUSION The three-miRNA panel (miR-29c-3p, miR-143-5p, and miR-205-5p) may become a novel non-invasive biological marker for nasopharyngeal cancer screening.
Collapse
Affiliation(s)
- Rongkang Li
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- The fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Chong Lu
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- The fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Weiqiang Yang
- Department of OtorhinolaryngologyPeking University Shenzhen HospitalShenzhenChina
| | - Yaqi Zhou
- Department of OtorhinolaryngologyPeking University Shenzhen HospitalShenzhenChina
| | - Jiatao Zhong
- Department of OtorhinolaryngologyPeking University Shenzhen HospitalShenzhenChina
| | - Xuan Chen
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Xinji Li
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Guocheng Huang
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Xiqi Peng
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Kaihao Liu
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- The fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Chunduo Zhang
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
| | - Hongyi Hu
- Department of OtorhinolaryngologyPeking University Shenzhen HospitalShenzhenChina
| | - Yongqing Lai
- Department of UrologyGuangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsPeking University Shenzhen HospitalClinical College of Anhui Medical UniversityShenzhenChina
- The fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| |
Collapse
|
19
|
Ruiz-Manriquez LM, Ledesma Pacheco SJ, Medina-Gomez D, Uriostegui-Pena AG, Estrada-Meza C, Bandyopadhyay A, Pathak S, Banerjee A, Chakraborty S, Srivastava A, Paul S. A Brief Review on the Regulatory Roles of MicroRNAs in Cystic Diseases and Their Use as Potential Biomarkers. Genes (Basel) 2022; 13:genes13020191. [PMID: 35205236 PMCID: PMC8872411 DOI: 10.3390/genes13020191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 02/04/2023] Open
Abstract
miRNAs are small endogenous conserved non-coding RNA molecules that regulate post-transcriptional gene expression through mRNA degradation or translational inhibition, modulating nearly 60% of human genes. Cystic diseases are characterized by the presence of abnormal fluid-filled sacs in the body, and though most cysts are benign, they can grow inside tumors and turn malignant. Recent evidence has revealed that the aberrant expression of a number of miRNAs present in extracellular fluids, including plasma or serum, urine, saliva, follicular fluid, and semen, contribute to different cystic pathologies. This review aims to describe the role of different miRNAs in three worldwide relevant cystic diseases: polycystic ovarian syndrome (PCOS), polycystic kidney disease (PKD), and pancreatic cyst tumors (PCTs), as well as their potential use as novel biomarkers.
Collapse
Affiliation(s)
- Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
| | - Schoenstatt Janin Ledesma Pacheco
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
| | - Daniel Medina-Gomez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
| | - Andrea G. Uriostegui-Pena
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
| | - Carolina Estrada-Meza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
| | - Anindya Bandyopadhyay
- C4 Rice Center, International Rice Research Institute, Manila 4031, Philippines;
- Synthetic Biology, Biofuel and Genome Editing R&D, Reliance Industries Ltd., Navi Mumbai 400701, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Faculty of Allied Health Sciences, Chennai 603103, India; (S.P.); (A.B.)
| | - Antara Banerjee
- Department of Medical Biotechnology, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Faculty of Allied Health Sciences, Chennai 603103, India; (S.P.); (A.B.)
| | - Samik Chakraborty
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Aashish Srivastava
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway;
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, Queretaro 76130, Mexico; (L.M.R.-M.); (S.J.L.P.); (D.M.-G.); (A.G.U.-P.); (C.E.-M.)
- Correspondence:
| |
Collapse
|
20
|
A Brief Review on the Regulatory Roles of MicroRNAs in Cystic Diseases and Their Use as Potential Biomarkers. Genes (Basel) 2022; 13:191. [PMID: 35205236 PMCID: PMC8872411 DOI: 10.3390/genes13020191&set/a 867452130+949943291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
miRNAs are small endogenous conserved non-coding RNA molecules that regulate post-transcriptional gene expression through mRNA degradation or translational inhibition, modulating nearly 60% of human genes. Cystic diseases are characterized by the presence of abnormal fluid-filled sacs in the body, and though most cysts are benign, they can grow inside tumors and turn malignant. Recent evidence has revealed that the aberrant expression of a number of miRNAs present in extracellular fluids, including plasma or serum, urine, saliva, follicular fluid, and semen, contribute to different cystic pathologies. This review aims to describe the role of different miRNAs in three worldwide relevant cystic diseases: polycystic ovarian syndrome (PCOS), polycystic kidney disease (PKD), and pancreatic cyst tumors (PCTs), as well as their potential use as novel biomarkers.
Collapse
|
21
|
A Brief Review on the Regulatory Roles of MicroRNAs in Cystic Diseases and Their Use as Potential Biomarkers. Genes (Basel) 2022. [DOI: 10.3390/genes13020191
expr 889616206 + 938882164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
miRNAs are small endogenous conserved non-coding RNA molecules that regulate post-transcriptional gene expression through mRNA degradation or translational inhibition, modulating nearly 60% of human genes. Cystic diseases are characterized by the presence of abnormal fluid-filled sacs in the body, and though most cysts are benign, they can grow inside tumors and turn malignant. Recent evidence has revealed that the aberrant expression of a number of miRNAs present in extracellular fluids, including plasma or serum, urine, saliva, follicular fluid, and semen, contribute to different cystic pathologies. This review aims to describe the role of different miRNAs in three worldwide relevant cystic diseases: polycystic ovarian syndrome (PCOS), polycystic kidney disease (PKD), and pancreatic cyst tumors (PCTs), as well as their potential use as novel biomarkers.
Collapse
|
22
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 507] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
23
|
Cao JM, Wang N, Hou SY, Qi X, Xiong W. Epigenetics effect on pathogenesis of thyroid-associated ophthalmopathy. Int J Ophthalmol 2021; 14:1441-1448. [PMID: 34540623 DOI: 10.18240/ijo.2021.09.22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Thyroid-associated ophthalmopathy (TAO) is an autoimmune disease. Recent studies have found the aberrant epigenetics in TAO, including DNA methylation, non-coding RNAs, and histone modification. Many genes have an aberrant level of methylation in TAO. For example, higher levels are found in CD14, MBP, ANGLE1, LYAR and lower levels in DRD4 and BOLL. Non-coding RNAs are involved in the immune response (miR-146a, miR-155, miR-96, miR-183), fibrosis regulation (miR-146a, miR-21, miR-29), adipogenesis (miR-27) and are thought to play roles in TAO. MicroRNA is also related to the clinical activity score (miR-Let7d-5p) and may be a predictor of glucocorticoid therapy (miR-224-5p). The quantities of H4 in TAO are increased compared with euthyroid control subjects, and the role of histone modifications in Graves' disease may lead to better understanding of its role in TAO. More studies are needed to explain the role of epigenetics in TAO and provide potential therapeutic strategies.
Collapse
Affiliation(s)
- Jia-Min Cao
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
| | - Nuo Wang
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
| | - Shi-Ying Hou
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xin Qi
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Xiong
- Department of Ophthalmology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
24
|
Karamanos NK, Piperigkou Z, Passi A, Götte M, Rousselle P, Vlodavsky I. Extracellular matrix-based cancer targeting. Trends Mol Med 2021; 27:1000-1013. [PMID: 34389240 DOI: 10.1016/j.molmed.2021.07.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Tumor extracellular matrix (ECM) operates in a coordinated mode with cancer and stroma cells to evoke the multistep process of metastatic potential. The remodeled tumor-associated matrix provides a point for direct or complementary therapeutic targeting. Here, we cover and critically address the importance of ECM networks and their macromolecules in cancer. We focus on the roles of key structural and functional ECM components, and their degradation enzymes and extracellular vesicles, aiming at improving our understanding of the mechanisms contributing to tumor initiation, growth, and dissemination, and discuss potential new approaches for ECM-based therapeutic targeting and diagnosis.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece.
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS-Université Lyon 1, SFR BioSciences Gerland-Lyon Sud, 7 Passage du Vercors, Lyon, France
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
25
|
Onyeisi JOS, Greve B, Espinoza-Sánchez NA, Kiesel L, Lopes CC, Götte M. microRNA-140-3p modulates invasiveness, motility, and extracellular matrix adhesion of breast cancer cells by targeting syndecan-4. J Cell Biochem 2021; 122:1491-1505. [PMID: 34180077 DOI: 10.1002/jcb.30071] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/21/2022]
Abstract
Syndecan-4, a predicted target of the microRNA miR-140-3p, plays an important role in multiple steps of tumor progression and is the second most abundant heparan sulfate proteoglycan produced by breast carcinoma cell lines. To investigate the potential functional relationship of miR-140-3p and syndecan-4, MDA-MB-231, SKBR3, and MCF-7 breast cancer (BC) cells were transiently transfected with pre-miR-140-3p, syndecan-4 small interfering RNAJ, or control reagents, respectively. Altered cell behavior was monitored by adhesion, migration, and invasion chamber assays. Moreover, the prognostic value of syndecan-4 was assessed by Kaplan-Maier Plotter analysis of gene expression data from tumor samples of 4929 patients. High expression of syndecan-4 was associated with better relapse-free survival in the whole collective of BC patients, but correlated with a worse survival in the subgroup of estrogen receptor negative and estrogen/progesterone-receptor negative patients. miR-140-3p expression was associated with improved survival irrespective of hormone receptor status. miR-140-3p overexpression induced posttranscriptional downregulation of syndecan-4, as demonstrated by quantitative real-time PCR (qPCR), flow cytometry, and luciferase assays, resulting in decreased BC cell migration and matrigel invasiveness. Furthermore, miR-140-3p overexpression and syndecan-4 silencing increased the adhesion of BC to fibronectin and laminin. qPCR analysis demonstrated that syndecan-4 silencing leads to altered gene expression of adhesion-related molecules, such as fibronectin and focal adhesion kinase, as well as in the gene expression of the proinvasive factors matrix metalloproteinase 2 and heparanase (also known as HPSE). We conclude that syndecan-4 is a novel target of miR-140-3p that regulates BC cell invasiveness and cell-matrix interactions in the tumor microenvironment.
Collapse
Affiliation(s)
- Jessica Oyie Sousa Onyeisi
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Disciplina de Biologia Molecular, Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Nancy Adriana Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Carla Cristina Lopes
- Disciplina de Biologia Molecular, Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.,Departamento de Ciências Biológicas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
26
|
Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers (Basel) 2021; 13:1441. [PMID: 33809973 PMCID: PMC8005147 DOI: 10.3390/cancers13061441] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue functionality and integrity demand continuous changes in distribution of major components in the extracellular matrices (ECMs) under normal conditions aiming tissue homeostasis. Major matrix degrading proteolytic enzymes are matrix metalloproteinases (MMPs), plasminogen activators, atypical proteases such as intracellular cathepsins and glycolytic enzymes including heparanase and hyaluronidases. Matrix proteases evoke epithelial-to-mesenchymal transition (EMT) and regulate ECM turnover under normal procedures as well as cancer cell phenotype, motility, invasion, autophagy, angiogenesis and exosome formation through vital signaling cascades. ECM remodeling is also achieved by glycolytic enzymes that are essential for cancer cell survival, proliferation and tumor progression. In this article, the types of major matrix remodeling enzymes, their effects in cancer initiation, propagation and progression as well as their pharmacological targeting and ongoing clinical trials are presented and critically discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| |
Collapse
|
27
|
Zolota V, Tzelepi V, Piperigkou Z, Kourea H, Papakonstantinou E, Argentou MI, Karamanos NK. Epigenetic Alterations in Triple-Negative Breast Cancer-The Critical Role of Extracellular Matrix. Cancers (Basel) 2021; 13:cancers13040713. [PMID: 33572395 PMCID: PMC7916242 DOI: 10.3390/cancers13040713] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subgroup of breast cancer characterized by genomic complexity and therapeutic options limited to only standard chemotherapy. Although it has been suggested that stratifying TNBC patients by pathway-specific molecular alterations may predict benefit from specific therapeutic agents, application in routine clinical practice has not yet been established. There is a growing body of the literature supporting that epigenetic modifications comprised by DNA methylation, chromatin remodeling and non-coding RNAs play a fundamental role in TNBC pathogenesis. Extracellular matrix (ECM) is a highly dynamic 3D network of macromolecules with structural and cellular regulatory roles. Alterations in the expression of ECM components result in uncontrolled matrix remodeling, thus affecting its ability to regulate vital functions of cancer cells, including proliferation, migration, adhesion, invasion and epithelial-to-mesenchymal transition (EMT). Recent molecular data highlight the major role of tumor microenvironment and ECM alterations in TNBC and approaches for targeting tumor microenvironment have recently been recognized as potential therapeutic strategies. Notably, many of the ECM/EMT modifications in cancer are largely driven by epigenetic events, highlighting the pleiotropic effects of the epigenetic network in TNBC. This article presents and critically discusses the current knowledge on the epigenetic alterations correlated with TNBC pathogenesis, with emphasis on those associated with ECM/EMT modifications, their prognostic and predictive value and their use as therapeutic targets.
Collapse
Affiliation(s)
- Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
- Correspondence: ; Tel.: +30-0693613366
| | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; (Z.P.); (N.K.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 26110 Patras, Greece
| | - Helen Kourea
- Department of Pathology, School of Medicine, University of Patras, 26504 Rion, Greece; (V.T.); (H.K.)
| | - Efthymia Papakonstantinou
- Department of Gynecology and Obstetrics School of Medicine, University of Patras, 26504 Rion, Greece;
| | - Maria-Ioanna Argentou
- Department of Surgery, School of Medicine, University of Patras, 26504 Rion, Greece;
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; (Z.P.); (N.K.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 26110 Patras, Greece
| |
Collapse
|
28
|
Ansari MA, Thiruvengadam M, Farooqui Z, Rajakumar G, Sajid Jamal QM, Alzohairy MA, Almatroudi A, Alomary MN, Chung IM, Al-Suhaimi EA. Nanotechnology, in silico and endocrine-based strategy for delivering paclitaxel and miRNA: Prospects for the therapeutic management of breast cancer. Semin Cancer Biol 2021; 69:109-128. [PMID: 31891780 DOI: 10.1016/j.semcancer.2019.12.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/06/2019] [Accepted: 12/25/2019] [Indexed: 02/07/2023]
Abstract
Breast cancer is one of the most prevalent and reoccurring cancers and the second most common reason of death in women. Despite advancements in therapeutic strategies for breast cancer, early tumor recurrence and metastasis in patients indicate resistance to chemotherapeutic medicines, such as paclitaxel due to the abnormal expression of ER and EGF2 in breast cancer cells. Therefore, the development of alternatives to paclitaxel is urgently needed to overcome challenges involving drug resistance. An increasing number of studies has revealed miRNAs as novel natural alternative substances that play a crucial role in regulating several physiological processes and have a close, adverse association with several diseases, including breast cancer. Due to the therapeutic potential of miRNA and paclitaxel in cancer research, the current review focuses on the differential roles of various miRNAs in breast cancer development and treatment. miRNA delivery to a specific target site, the development of paclitaxel and miRNA formulations, and nanotechnological strategies for the delivery of nanopaclitaxel in the management of breast cancer are discussed. These strategies involve improving the cellular uptake and bioavailability and reducing the toxicity of free paclitaxel to achieve accumulation tumor site. Furthermore, a molecular docking study was performed to ascertain the enhanced anticancer activity of the nanoformulation of ANG1005 and Abraxane. An in silico analysis revealed that ANG1005 and Abraxane nanoformulations have superior and significantly enhanced interactions with the proteins α-tubulin and Bcl-2. Therefore, ANG1005 and Abraxane may be more suitable in the therapeutic management of breast cancer than the existing free paclitaxel. miRNAs can revert abnormal gene expression to normalcy; since miRNAs serve as tumor suppressors. Therefore, restoration of particular miRNAs levels as a replacement therapy may be an effective endocrine potential strategy for treating ER positive/ negative breast cancers.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Diseases Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Zeba Farooqui
- College of Pharmacy, University of Houston, Houston, TX, 77204, United States
| | - Govindaswamy Rajakumar
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al-Bukayriyah, Saudi Arabia
| | - Mohammad A Alzohairy
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Mohammad N Alomary
- National Center of Biotechnology, Life Science and Environmental Research Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh, Saudi Arabia
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea.
| | - Ebtesam Abdullah Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, Saudi Arabia; Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| |
Collapse
|
29
|
Sang Y, Kong P, Zhang S, Zhang L, Cao Y, Duan X, Sun T, Tao Z, Liu W. SGK1 in Human Cancer: Emerging Roles and Mechanisms. Front Oncol 2021; 10:608722. [PMID: 33542904 PMCID: PMC7851074 DOI: 10.3389/fonc.2020.608722] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Serum and glucocorticoid-induced protein kinase 1 (SGK1) is a member of the "AGC" subfamily of protein kinases, which shares structural and functional similarities with the AKT family of kinases and displays serine/threonine kinase activity. Aberrant expression of SGK1 has profound cellular consequences and is closely correlated with human cancer. SGK1 is considered a canonical factor affecting the expression and signal transduction of multiple genes involved in the genesis and development of many human cancers. Abnormal expression of SGK1 has been found in tissue and may hopefully become a useful indicator of cancer progression. In addition, SGK1 acts as a prognostic factor for cancer patient survival. This review systematically summarizes and discusses the role of SGK1 as a diagnostic and prognostic biomarker of diverse cancer types; focuses on its essential roles and functions in tumorigenesis, cancer cell proliferation, apoptosis, invasion, metastasis, autophagy, metabolism, and therapy resistance and in the tumor microenvironment; and finally summarizes the current understanding of the regulatory mechanisms of SGK1 at the molecular level. Taken together, this evidence highlights the crucial role of SGK1 in tumorigenesis and cancer progression, revealing why it has emerged as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Piaoping Kong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shizhen Zhang
- The Cancer Institute of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Cao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
30
|
Pibuel MA, Poodts D, Díaz M, Hajos SE, Lompardía SL. The scrambled story between hyaluronan and glioblastoma. J Biol Chem 2021; 296:100549. [PMID: 33744285 PMCID: PMC8050860 DOI: 10.1016/j.jbc.2021.100549] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in cancer biology are revealing the importance of the cancer cell microenvironment on tumorigenesis and cancer progression. Hyaluronan (HA), the main glycosaminoglycan in the extracellular matrix, has been associated with the progression of glioblastoma (GBM), the most frequent and lethal primary tumor in the central nervous system, for several decades. However, the mechanisms by which HA impacts GBM properties and processes have been difficult to elucidate. In this review, we provide a comprehensive assessment of the current knowledge on HA's effects on GBM biology, introducing its primary receptors CD44 and RHAMM and the plethora of relevant downstream signaling pathways that can scramble efforts to directly link HA activity to biological outcomes. We consider the complexities of studying an extracellular polymer and the different strategies used to try to capture its function, including 2D and 3D in vitro studies, patient samples, and in vivo models. Given that HA affects not only migration and invasion, but also cell proliferation, adherence, and chemoresistance, we highlight the potential role of HA as a therapeutic target. Finally, we review the different existing approaches to diminish its protumor effects, such as the use of 4-methylumbelliferone, HA oligomers, and hyaluronidases and encourage further research along these lines in order to improve the survival and quality of life of GBM patients.
Collapse
Affiliation(s)
- Matías Arturo Pibuel
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| | - Daniela Poodts
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Mariángeles Díaz
- Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvia Elvira Hajos
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina
| | - Silvina Laura Lompardía
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU)-CONICET, Universidad de Buenos Aires, Capital Federal, Argentina.
| |
Collapse
|
31
|
Kääriäinen A, Pesola V, Dittmann A, Kontio J, Koivunen J, Pihlajaniemi T, Izzi V. Machine Learning Identifies Robust Matrisome Markers and Regulatory Mechanisms in Cancer. Int J Mol Sci 2020; 21:E8837. [PMID: 33266472 PMCID: PMC7700160 DOI: 10.3390/ijms21228837] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
The expression and regulation of matrisome genes-the ensemble of extracellular matrix, ECM, ECM-associated proteins and regulators as well as cytokines, chemokines and growth factors-is of paramount importance for many biological processes and signals within the tumor microenvironment. The availability of large and diverse multi-omics data enables mapping and understanding of the regulatory circuitry governing the tumor matrisome to an unprecedented level, though such a volume of information requires robust approaches to data analysis and integration. In this study, we show that combining Pan-Cancer expression data from The Cancer Genome Atlas (TCGA) with genomics, epigenomics and microenvironmental features from TCGA and other sources enables the identification of "landmark" matrisome genes and machine learning-based reconstruction of their regulatory networks in 74 clinical and molecular subtypes of human cancers and approx. 6700 patients. These results, enriched for prognostic genes and cross-validated markers at the protein level, unravel the role of genetic and epigenetic programs in governing the tumor matrisome and allow the prioritization of tumor-specific matrisome genes (and their regulators) for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Anni Kääriäinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Vilma Pesola
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Annalena Dittmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Juho Kontio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland; (A.K.); (V.P.); (A.D.); (J.K.); (J.K.); (T.P.)
- Faculty of Medicine, University of Oulu, P.O. BOX 8000, FI-90014 Oulu, Finland
- Finnish Cancer Institute, 00130 Helsinki, Finland
| |
Collapse
|
32
|
Huang G, Li H, Wang J, Peng X, Liu K, Zhao L, Zhang C, Chen X, Lai Y, Ni L. Combination of tumor suppressor miR-20b-5p, miR-30a-5p, and miR-196a-5p as a serum diagnostic panel for renal cell carcinoma. Pathol Res Pract 2020; 216:153152. [PMID: 32823234 DOI: 10.1016/j.prp.2020.153152] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Renal cell carcinoma (RCC) accounts for 3 % of cancer patients. Early detection influences the therapeutic strategy and significantly improves patients' survival rates. Stable existing circulating miRNAs could be a promising diagnostic biomarker. METHODS Previously our team demonstrated the anti-tumor effect of miR-20b-5p, miR-30a-5p and miR-196a-5p in RCC tissue and cell lines. Here, based on 110 RCC patients and 110 health control, we investigated serum expression of these three miRNAs in the testing set and the validation set separately by using quantitative real-time PCR. A three-miRNA panel with high diagnostic efficiency was constructed. Correlations between these miRNAs and clinical parameters were investigated. Additionally, the TCGA dataset and bioinformatic analysis are used for the functional exploration of these miRNAs. RESULTS Serum expression levels of miR-20b-5p, miR-30a-5p were significantly reduced in RCC patients, while miR-196a-5p expression level was up-regulated (p < 0.001). miR-20b-5p, miR-30a-5p and miR-196a-5p had moderate diagnostic ability for RCC (AUC = 0.807, 0.766 and 0.719 in the testing set, respectively). The AUC of the three-miRNA panel was 0.949 in the testing set and 0.938 in the validation set. Specifically, the serum expression level of miR-196a-5p was significantly down-regulated in RCC patients with higher Fuhrman grade (p = 0.051). TCGA dataset analysis showed that the three-miRNA panel probably participated in RCC by targeting ITGA4 and NRP2. CONCLUSION The three-miRNA panel could serve as a promising non-invasive biomarker for RCC detection.
Collapse
Affiliation(s)
- Guocheng Huang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Huijuan Li
- Department of Urology, Shandong Provincial Hospital, Jinan, Shandong, 250021, China
| | - Jingyao Wang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xiqi Peng
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Kaihao Liu
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Anhui Medical University, Hefei, Anhui, 230032, China
| | - Liwen Zhao
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Anhui Medical University, Hefei, Anhui, 230032, China
| | - Chunduo Zhang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xuan Chen
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Yongqing Lai
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shantou University Medical College, Shantou, Guangdong, 515041, China.
| | - Liangchao Ni
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shantou University Medical College, Shantou, Guangdong, 515041, China.
| |
Collapse
|
33
|
Li T, Mao C, Wang X, Shi Y, Tao Y. Epigenetic crosstalk between hypoxia and tumor driven by HIF regulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:224. [PMID: 33109235 PMCID: PMC7592369 DOI: 10.1186/s13046-020-01733-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Hypoxia is the major influence factor in physiological and pathological courses which are mainly mediated by hypoxia-inducible factors (HIFs) in response to low oxygen tensions within solid tumors. Under normoxia, HIF signaling pathway is inhibited due to HIF-α subunits degradation. However, in hypoxic conditions, HIF-α is activated and stabilized, and HIF target genes are successively activated, resulting in a series of tumour-specific activities. The activation of HIFs, including HIF-1α, HIF-2α and HIF-3α, subsequently induce downstream target genes which leads to series of responses, the resulting abnormal processes or metabolites in turn affect HIFs stability. Given its functions in tumors progression, HIFs have been regarded as therapeutic targets for improved treatment efficacy. Epigenetics refers to alterations in gene expression that are stable between cell divisions, and sometimes between generations, but do not involve changes in the underlying DNA sequence of the organism. And with the development of research, epigenetic regulation has been found to play an important role in the development of tumors, which providing accumulating basic or clinical evidences for tumor treatments. Here, given how little has been reported about the overall association between hypoxic tumors and epigenetics, we made a more systematic review from epigenetic perspective in hope of helping others better understand hypoxia or HIF pathway, and providing more established and potential therapeutic strategies in tumors to facilitate epigenetic studies of tumors.
Collapse
Affiliation(s)
- Tiansheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chao Mao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ying Shi
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Yongguang Tao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
34
|
Mao S, Lu Z, Zheng S, Zhang H, Zhang G, Wang F, Huang J, Lei Y, Wang X, Liu C, Sun N, He J. Exosomal miR-141 promotes tumor angiogenesis via KLF12 in small cell lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:193. [PMID: 32958011 PMCID: PMC7504642 DOI: 10.1186/s13046-020-01680-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/17/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Angiogenesis, a basic requirement for tumor cell survival, is considered to be a malignant characteristic of small cell lung cancer (SCLC) and is closely related to the poor outcomes of SCLC patients. miR-141 has been found to play pro- and antiangiogenic roles in different cancers, but its role in SCLC angiogenesis has never been explored. METHODS Total RNA was isolated from plasm exosomes and serum of SCLC patients to examine the expression of miR-141 by qRT-PCR. Cell proliferation, invasion, migration, tube formation assay, aortic ring assay and mouse tumor model were used to investigate the effect of exosomal miR-141 in angiogenesis in vitro and in vivo. Dual-luciferase assay was conducted to explore the target gene of miR-141. RESULTS Circulating miR-141 was upregulated in samples from 122 SCLC patients compared with those from normal volunteers and that the increase in miR-141 was significantly associated with advanced TNM stages, implying the potential oncogenic role of miR-141 in SCLC malignancy. In vitro, miR-141 that was packaged into SCLC cell-secreted exosomes and delivered to human umbilical vein vascular endothelial cells (HUVECs) via exosomes facilitated HUVEC proliferation, invasion, migration and tube formation and promoted microvessel sprouting from mouse aortic rings. Matrigel plug assays demonstrated that SCLC cell-derived exosomal miR-141 induced neoangiogenesis in vivo. Furthermore, mouse subcutaneous tumor nodules that were developed from miR-141-overexpressing SCLC cells had a higher microvessel density (MVD) and grew faster than those developed from negative control cells. KLF12 was found to be the direct target gene of miR-141 and that the proangiogenic effect of miR-141 on HUVECs was abrogated by KLF12 overexpression. CONCLUSIONS Our results demonstrate the specific function of the exosomal miR-141/KLF12 pathway in SCLC angiogenesis for the first time and provide potential novel targets for antiangiogenic therapies for SCLC patients.
Collapse
Affiliation(s)
- Shuangshuang Mao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiliang Lu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Sufei Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Feng Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianbing Huang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinfeng Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chengming Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
35
|
Shi M, Zong X, Chen L, Guo X, Ding X. MiR-506-3p regulates autophagy and proliferation in post-burn skin fibroblasts through post-transcriptionally suppressing Beclin-1 expression. In Vitro Cell Dev Biol Anim 2020; 56:522-532. [PMID: 32754856 DOI: 10.1007/s11626-020-00472-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) is involved in diverse biological processes of cells including dermal fibroblasts that contributed to wound healing and resulted in keloid scarring. MiR-506-3p has been identified as a tumor suppressor or oncogene in fibroblasts of various cancers, while the role of miR-506-3p in regulating functions of post-burn dermal fibroblasts is poorly known. In this study, miR-506-3p was confirmed to be significantly downregulated in burned tissues and heat-stimulated dermal fibroblasts. Expression levels of autophagy-related proteins suggested thermal stimulus promoting the autophagy in dermal fibroblasts. Then, miR-506-3p inhibition enhanced cell proliferation and cell cycle process in dermal fibroblasts after thermal stimulus, whereas overexpression of miR-506-3p showed the opposite effect. Western blot assay showed that inhibition of miR-506-3p resulted in the upregulation of the expression levels of LC3-II, ATG5, and structural protein collagen I, as well as the downregulation of p62. Marker proteins of intermolecular cross-links in collagen synthesis, including hydroxylysylpyridinoline (HP), lysinepyridine (LP), and lysyl hydroxylase 2 (LH2), were increased by miR-506-3p overexpression and decreased by miR-506-3p inhibition. Moreover, transfection with miR-506-3p mimic suppressed the proliferation and autophagy in heat-stimulated dermal fibroblasts in a dose-dependent manner. Subsequently, dual luciferase reporter gene assay demonstrated that Beclin-1 was a direct target of miR-506-3p, and reintroduction of Beclin-1 could antagonize the suppressive effect of miR-506-3p overexpression on fibroblast proliferation, autophagy, and the intermolecular cross-links in collagen synthesis. Taken together, our findings showed that miR-506-3p regulated autophagy and proliferation in post-burn skin fibroblasts through post-transcriptionally suppressing Beclin-1 expression.
Collapse
Affiliation(s)
- Min Shi
- School of Medicine, Xi'an Peihua University, Xi'an, China
| | - Xiaoming Zong
- Department of Oncology, Shaanxi Friendship Hospital, Xi'an, China
| | - Lei Chen
- School of Medicine, Xi'an Peihua University, Xi'an, China
| | - Xiaobo Guo
- Department of Hematology, Xi'an Central Hospital, Xi'an, China
| | - Xinqiang Ding
- Department of Dermatology, Xi'an Children's Hospital, 69 Xijuyuan Road, Lianhu District, Xi'an, 710000, China.
| |
Collapse
|
36
|
Karamanos NK. Extracellular matrix: key structural and functional meshwork in health and disease. FEBS J 2020; 286:2826-2829. [PMID: 31379113 DOI: 10.1111/febs.14992] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) is a multifunctional 3D network of interconnected macromolecules that mediate cellular responses in normal and pathological conditions, including wound healing, tissue homeostasis, skeletal and cardiovascular disease, cancer and drug resistance. The potential of targeting ECM components, such as proteoglycans and glycosaminoglycans, matrix-degrading enzymes and miRNAs will serve as a novel tool for innovative diagnostic methods and therapeutic strategies. With this Special Issue on Extracellular Matrix in Health and Disease, The FEBS Journal offers an updated overview of the functional properties and biological roles of several ECM components, highlighting the significance of ECM targeting in disease management.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
37
|
Ge Y, Wang Q, Shao W, Zhao Y, Shi Q, Yuan Q, Cui L. Circulating let-7f-5p improve risk prediction of prostate cancer in patients with benign prostatic hyperplasia. J Cancer 2020; 11:4542-4549. [PMID: 32489471 PMCID: PMC7255360 DOI: 10.7150/jca.45077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Although the prostate-specific antigen (PSA) testing was widely used for early detection of prostate cancer (PCa), it is difficult for PSA to distinguish the PCa from benign prostatic hyperplasia (BPH) patients. Emerging evidence has shown that microRNA (miRNA) was a promising biomarker for PCa screening. Methods: We applied miRNA profiling from microarray or high-throughput sequencing in Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases to identify the differentially expressed miRNAs in PCa patients (n = 1,017) and controls (n = 413). Then, qRT-PCR analysis was used to validate the expression of candidate miRNAs in our independent cohort, include 66 PCa cases and 63 BPH patients diagnosed by biopsy. The area under the receiver operating characteristic curve (AUC) was conducted to evaluate the diagnostic efficacy of miRNAs and PSA. Results: In the microarray analysis, we identified two consistently differently expressed miRNAs (miR-103a-3p and let-7f-5p) between PCa patients and controls. In the subsequent qRT-PCR analysis, the let-7f-5p was upregulated in PCa compared with BPH patients (P=2.17E-07), but no statistically difference of miR-103a-3p expression was observed (P=0.456). The AUC was 0.904 for combination of lef-7f-5p and PSA, which was significantly higher than that of let-7f-5p (0.782) or PSA (0.795) alone (P=7.55E-04 and P=2.09E-03, respectively). Besides, the results of decision curve analysis and nomogram prediction indicated that combination of let-7f-5p and PSA had superior predictive accuracy of PCa. Conclusions: Our study suggests that plasma let-7f-5p combining PSA could serve as potentially diagnostic biomarkers for PCa.
Collapse
Affiliation(s)
- Yuqiu Ge
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qiangdong Wang
- Department of Urology, Huaiyin Hospital of Huai'an City, Huai'an, China.,Department of Urology, Huaiyin People's Hospital of Huai'an City, Huai'an, China
| | - Wei Shao
- Department of Science and Technology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - You Zhao
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianqian Shi
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qinbo Yuan
- Department of Urology, Huaiyin Hospital of Huai'an City, Huai'an, China.,Department of Urology, Huaiyin People's Hospital of Huai'an City, Huai'an, China
| | - Li Cui
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
38
|
Iozzo RV, Theocharis AD, Neill T, Karamanos NK. Complexity of matrix phenotypes. Matrix Biol Plus 2020; 6-7:100038. [PMID: 33543032 PMCID: PMC7852209 DOI: 10.1016/j.mbplus.2020.100038] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix is engaged in an ever-evolving and elegant ballet of dynamic reciprocity that directly and bi-directionally regulates cell behavior. Homeostatic and pathophysiological changes in cell-matrix signaling cascades manifest as complex matrix phenotypes. Indeed, the extracellular matrix can be implicated in virtually every known human disease, thus, making it the most critical and dynamic "organ" in the human body. The overall goal of this Special Issue is to provide an accurate and inclusive functional definition that addresses the inherent complexity of matrix phenotypes. This goal is summarily achieved via a corpus of expertly written articles, reviews and original research, focused at answering this question empirically and fundamentally via state-of-the-art methods and research strategies.
Collapse
Key Words
- ADAM, a disintegrin and metalloproteinases
- AGE, advanced glycation end products
- Angiogenesis
- Cancer
- Collagen
- DDR1, discoidin domain receptor 1
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- EMILIN1, elastin microfibril interfacer 1
- EMILIN2, elastin microfibril interfacer 2
- EMT, epithelial-mesenchymal transition
- ERα, estrogen receptor α
- ERβ, estrogen receptor β
- GBM, glioblastoma
- HA, hyaluronan
- HAS2, hyaluronan synthase 2
- HAS2-AS1, HAS2 antisense 1
- HB-EGF, heparin binding EGF
- HMGA2, high-mobility group AT-Hook 2
- IBC, inflammatory breast cancer
- IGF-IR, insulin growth factor I receptor
- IR-A, insulin receptor A
- LEKTI, lympho-epithelial Kazal-type inhibitor
- LOX, lysyl oxidases
- LTBP, latent TGFβ-binding proteins
- MAGP, microfibril-associated glycoproteins
- MET, mesenchymal-epithelial transition
- MMP, matrix metalloproteinases
- Methodologies
- OB, osteoblast
- OI, osteogenesis imperfecta
- PARs, protease activated receptors
- PG, proteoglycans
- PLL, poly-l-lysine
- Proteoglycans
- ROS, reactive oxygen species
- RTK, receptor tyrosine kinase
- SLRP, small leucine rich proteoglycans
- SSR, solar-simulated radiation
- TGFβ, transforming growth factor β
- TNT, tunneling nanotubes
- UVR, ultraviolet radiation
- VEGF, vascular endothelial growth factor
- miR, microRNA
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
Collapse
Affiliation(s)
- Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
39
|
Piperigkou Z, Franchi M, Riethmüller C, Götte M, Karamanos NK. miR-200b restrains EMT and aggressiveness and regulates matrix composition depending on ER status and signaling in mammary cancer. Matrix Biol Plus 2020; 6-7:100024. [PMID: 33543022 PMCID: PMC7852204 DOI: 10.1016/j.mbplus.2020.100024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Secreted microRNAs (miRNAs) reside in a complex regulatory network with extracellular matrix (ECM) macromolecules, which affect cell-cell communication, therefore miRNA expression highlights its significance in several aspects of human diseases, including cancer. miRNA-mediated regulation of breast cancer has received considerable attention due to evidence that shows miRNAs to mediate estrogen receptor (ER) status, metastasis, chemoresistance and epithelial-to-mesenchymal transition (EMT). miR-200b is a pluripotent miRNA, which is inversely regulated by ERα and ERβ in mammary cancer. It has been identified as tumor suppressor and EMT inhibitor serving as a critical biomarker, as its expression in breast tumor determines the disease-free survival, thus highlighting its roles in breast cancer invasion and metastasis. The main goal of this study was to investigate the role of miR-200b in modulating the behavior of breast cancer cells with different ER status. We demonstrate that estrogen signaling through ERs reduces miR-200b expression levels in ERα-positive breast cancer cells. Moreover, miR-200b upregulation reduces the aggressive phenotype of ERβ-positive breast cancer cells by inhibiting cell invasiveness and motility, followed by ECM reorganization as well as cytoskeletal and morphological changes concluded from deep inspection of cell topography. Future investigation towards the mechanistic perspective of miR-200b effects in the behavior of aggressive mammary cancer cells appears rewarding in order to expand our understanding of miR-200b as a novel mediator beyond breast cancer diagnosis and pharmaceutical targeting.
Collapse
Key Words
- Breast cancer
- ECM, extracellular matrix
- EGFR, epidermal growth factor receptor
- EMT, epithelial-to-mesenchymal-transition
- ER, estrogen receptor
- Erk, extracellular signal-regulated kinase
- Estrogen receptors
- Extracellular matrix
- GAG, glycosaminoglycan
- GF, growth factor
- HER2, human epidermal growth factor receptor 2
- IGF-IR, insulin-like growth factor receptor type I
- IL, interleukin
- MMP, matrix metalloproteinase
- PG, proteoglycan
- PR, progesterone receptor
- RISC, RNA-induced silencing complex
- SERM, selective estrogen receptor modulator
- TGFβ, transforming growth factor beta
- miR-200b
- miRNA, microRNA
- miRNAs
- pre-miRNA, precursor miRNA
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | | | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
40
|
Gulberti S, Mao X, Bui C, Fournel-Gigleux S. The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs). Semin Cancer Biol 2020; 62:68-85. [DOI: 10.1016/j.semcancer.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
|
41
|
Circulating Small Noncoding RNAs Have Specific Expression Patterns in Plasma and Extracellular Vesicles in Myelodysplastic Syndromes and Are Predictive of Patient Outcome. Cells 2020; 9:cells9040794. [PMID: 32224889 PMCID: PMC7226126 DOI: 10.3390/cells9040794] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are hematopoietic stem cell disorders with large heterogeneity at the clinical and molecular levels. As diagnostic procedures shift from bone marrow biopsies towards less invasive techniques, circulating small noncoding RNAs (sncRNAs) have become of particular interest as potential novel noninvasive biomarkers of the disease. We aimed to characterize the expression profiles of circulating sncRNAs of MDS patients and to search for specific RNAs applicable as potential biomarkers. We performed small RNA-seq in paired samples of total plasma and plasma-derived extracellular vesicles (EVs) obtained from 42 patients and 17 healthy controls and analyzed the data with respect to the stage of the disease, patient survival, response to azacitidine, mutational status, and RNA editing. Significantly higher amounts of RNA material and a striking imbalance in RNA content between plasma and EVs (more than 400 significantly deregulated sncRNAs) were found in MDS patients compared to healthy controls. Moreover, the RNA content of EV cargo was more homogeneous than that of total plasma, and different RNAs were deregulated in these two types of material. Differential expression analyses identified that many hematopoiesis-related miRNAs (e.g., miR-34a, miR-125a, and miR-150) were significantly increased in MDS and that miRNAs clustered on 14q32 were specifically increased in early MDS. Only low numbers of circulating sncRNAs were significantly associated with somatic mutations in the SF3B1 or DNMT3A genes. Survival analysis defined a signature of four sncRNAs (miR-1237-3p, U33, hsa_piR_019420, and miR-548av-5p measured in EVs) as the most significantly associated with overall survival (HR = 5.866, p < 0.001). In total plasma, we identified five circulating miRNAs (miR-423-5p, miR-126-3p, miR-151a-3p, miR-125a-5p, and miR-199a-3p) whose combined expression levels could predict the response to azacitidine treatment. In conclusion, our data demonstrate that circulating sncRNAs show specific patterns in MDS and that their expression changes during disease progression, providing a rationale for the potential clinical usefulness of circulating sncRNAs in MDS prognosis. However, monitoring sncRNA levels in total plasma or in the EV fraction does not reflect one another, instead, they seem to represent distinctive snapshots of the disease and the data should be interpreted circumspectly with respect to the type of material analyzed.
Collapse
|
42
|
Gao Y, Wu P, Ma Y, Xue Y, Liu Y, Zheng J, Liu X, He Q, Ma J, Liu L, Wang P. Circular RNA USP1 regulates the permeability of blood-tumour barrier via miR-194-5p/FLI1 axis. J Cell Mol Med 2019; 24:342-355. [PMID: 31654502 PMCID: PMC6933377 DOI: 10.1111/jcmm.14735] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/21/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
Recent studies indicate circular RNAs are related to dysregulation of vascular endothelial cell function, yet the underlying mechanisms have remained elusive. Here, we characterized the functional role of circular RNA USP1 (circ‐USP1) in the regulation of the blood‐tumour barrier (BTB) permeability and the potential mechanisms. In the current study, the circ‐USP1 expressing level was up‐regulated in glioma cerebral microvascular endothelial cells (GECs) of the BTB model in vitro. Knockdown of circ‐USP1 disrupted the barrier integrity, increased its permeability as well as reduced tight junction‐related protein claudin‐5, occludin and ZO‐1 expressions in GECs. Bioinformatic prediction and luciferase assay indicated that circ‐USP1 bound to miR‐194‐5p and suppressed its activity. MiR‐194‐5p contributed to circ‐USP1 knockdown‐induced increase of BTB permeability via targeting and down‐regulating transcription factor FLI1. Furthermore, FLI1 regulated the expressions of claudin‐5, occludin and ZO‐1 in GECs through binding to their promoter regions. Single or combined treatment of circ‐USP1 and miR‐194‐5p effectively promoted anti‐tumour drug doxorubicin across BTB to induce apoptosis of glioma cells. Overall, this present study identified the crucial regulation of circ‐USP1 on BTB permeability via miR‐194‐5p/FLI1 axis‐mediated regulation of tight junction proteins, which might facilitate the development of therapeutics against human gliomas.
Collapse
Affiliation(s)
- Yang Gao
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China.,Laboratory of Digital Health, Medaxis Technology Co., Ltd., Chengdu, China
| | - Peiqi Wu
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yawen Ma
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China
| | - Yixue Xue
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang, China
| | - Qianru He
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Jun Ma
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Libo Liu
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Ping Wang
- Department of Neurobiology, College of Life Science, China Medical University, Shenyang, China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| |
Collapse
|