1
|
Dan Y, Zhao X, Li J, Zhong H, Zhang H, Wu J, He J, Li L, Song Q, Xu B. Harnessing pseudogenes for lung cancer: A novel epigenetic target in diagnosis, prognosis and treatment. Crit Rev Oncol Hematol 2025; 208:104645. [PMID: 39900316 DOI: 10.1016/j.critrevonc.2025.104645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
Pseudogenes are abundantly present in the human genome and are often thought of as nonfunctional nucleotide sequences, but a growing body of research suggests that pseudogenes can play important biological roles through a variety of pathways, and can be involved in the development of cancer. Lung cancer is one of the most prevalent cancers in the world and it is crucial to find new therapeutic strategies for the treatment of lung cancer. In recent years, studies on the effects of pseudogenes on lung carcinogenesis have increased rapidly. This has pointed to new directions in the diagnosis and treatment of lung cancer. Aim of this paper is to comprehensively discuss the role and influence of pseudogenes in the lung cancer, and the potential of pseudogenes as novel epigenetic targets in lung cancer diagnosis and prognosis and treatment, which is significant for realizing the clinical benefits of pseudogenes.
Collapse
Affiliation(s)
- Yuchao Dan
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Xinyi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Jing Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Hao Zhong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Haohan Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Junju He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
2
|
Guo L, Zhang P, Zhang M, Liang P, Zhou S. LncRNA AGAP2-AS1 stabilizes ATG9A to promote autophagy in endothelial cells - Implications for burn wound healing. Exp Cell Res 2024; 443:114310. [PMID: 39481796 DOI: 10.1016/j.yexcr.2024.114310] [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: 05/06/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Deep second- or mixed-degree burn lesions are difficult to heal due to the impaired dermis supporting of epidermis renewal and nutrition delivery. Early dermis debridement and preservation speed healing and enhance results, emphasizing the need of knowing processes that promote burn-denatured dermis recovery, notably endothelial cell angiogenesis and autophagy. Integrative bioinformatics investigations identified AGAP2-AS1 as a highly elevated lncRNA in burn tissues. Pearson's correlation study connected AGAP2-AS1 to 112 differently co-expressed protein-coding genes involved in burn healing processes such cell cycle and TGF-beta receptor signaling. Experimental validation showed that heat damage elevated AGAP2-AS1 in HUVECs and HDMECs. Functionally, AGAP2-AS1 overexpression in heat-denatured HUVECs and HDMECs increased cell survival, migration, invasion, and angiogenesis. In addition, AGAP2-AS1 overexpression increased endothelial cell autophagy. Additional investigation showed AGAP2-AS1's association with ATG9A, stabilizing it. Post-heat damage, ATG9A knockdown drastically reduced HUVEC and HDMEC survival, migration, invasion, angiogenesis, and autophagy. More notably, ATG9A knockdown drastically reduced the benefits of AGAP2-AS1 overexpression on endothelial cell functions and autophagy. The positive association between AGAP2-AS1 and ATG9A expression in burn tissue samples highlights their crucial roles in endothelial cell response to heat injury, indicating that targeting this axis may aid burn wound healing. The research found that lncRNA AGAP2-AS1 stabilizes ATG9A and promotes autophagy in endothelial cells. These results imply that targeting the AGAP2-AS1/ATG9A axis may improve angiogenesis and tissue regeneration in burn injuries, revealing burn wound healing molecular pathways.
Collapse
Affiliation(s)
- Le Guo
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Minghua Zhang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pengfei Liang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Situo Zhou
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
3
|
Gao H, Li Z, Gan L, Chen X. The Role and Potential Mechanisms of Rehabilitation Exercise Improving Cardiac Remodeling. J Cardiovasc Transl Res 2024; 17:923-934. [PMID: 38558377 DOI: 10.1007/s12265-024-10498-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/08/2024] [Indexed: 04/04/2024]
Abstract
Rehabilitation exercise is a crucial non-pharmacological intervention for the secondary prevention and treatment of cardiovascular diseases, effectively ameliorating cardiac remodeling in patients. Exercise training can mitigate cardiomyocyte apoptosis, reduce extracellular matrix deposition and fibrosis, promote angiogenesis, and regulate inflammatory response to improve cardiac remodeling. This article presents a comprehensive review of recent research progress, summarizing the pivotal role and underlying mechanism of rehabilitation exercise in improving cardiac remodeling and providing valuable insights for devising effective rehabilitation treatment programs. Graphical Abstract.
Collapse
Affiliation(s)
- Haizhu Gao
- Colleague of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhongxin Li
- Colleague of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Lijun Gan
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, No.89 Guhuai Road, Jining, 272029, Shandong, China
| | - Xueying Chen
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, No.89 Guhuai Road, Jining, 272029, Shandong, China.
- Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
4
|
Ding Y, Huang X, Ji T, Qi C, Gao X, Wei R. The emerging roles of miRNA-mediated autophagy in ovarian cancer. Cell Death Dis 2024; 15:314. [PMID: 38702325 PMCID: PMC11068799 DOI: 10.1038/s41419-024-06677-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/06/2024]
Abstract
Ovarian cancer is one of the common tumors of the female reproductive organs. It has a high mortality rate, is highly heterogeneous, and early detection and primary prevention are very complex. Autophagy is a cellular process in which cytoplasmic substrates are targeted for degradation in lysosomes through membrane structures called autophagosomes. The periodic elimination of damaged, aged, and redundant cellular molecules or organelles through the sequential translation between amino acids and proteins by two biological processes, protein synthesis, and autophagic protein degradation, helps maintain cellular homeostasis. A growing number of studies have found that autophagy plays a key regulatory role in ovarian cancer. Interestingly, microRNAs regulate gene expression at the posttranscriptional level and thus can regulate the development and progression of ovarian cancer through the regulation of autophagy in ovarian cancer. Certain miRNAs have recently emerged as important regulators of autophagy-related gene expression in cancer cells. Moreover, miRNA analysis studies have now identified a sea of aberrantly expressed miRNAs in ovarian cancer tissues that can affect autophagy in ovarian cancer cells. In addition, miRNAs in plasma and stromal cells in tumor patients can affect the expression of autophagy-related genes and can be used as biomarkers of ovarian cancer progression. This review focuses on the potential significance of miRNA-regulated autophagy in the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yamin Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Xuan Huang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Tuo Ji
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, China
| | - Cong Qi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Xuzhu Gao
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, China.
| | - Rongbin Wei
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China.
| |
Collapse
|
5
|
Ateeq M, Broadwin M, Sellke FW, Abid MR. Extracellular Vesicles' Role in Angiogenesis and Altering Angiogenic Signaling. Med Sci (Basel) 2024; 12:4. [PMID: 38249080 PMCID: PMC10801520 DOI: 10.3390/medsci12010004] [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: 11/16/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Angiogenesis, the process of new blood vessels formation from existing vasculature, plays a vital role in development, wound healing, and various pathophysiological conditions. In recent years, extracellular vesicles (EVs) have emerged as crucial mediators in intercellular communication and have gained significant attention for their role in modulating angiogenic processes. This review explores the multifaceted role of EVs in angiogenesis and their capacity to modulate angiogenic signaling pathways. Through comprehensive analysis of a vast body of literature, this review highlights the potential of utilizing EVs as therapeutic tools to modulate angiogenesis for both physiological and pathological purposes. A good understanding of these concepts holds promise for the development of novel therapeutic interventions targeting angiogenesis-related disorders.
Collapse
Affiliation(s)
- Maryam Ateeq
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (M.A.); (M.B.); (F.W.S.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (M.A.); (M.B.); (F.W.S.)
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (M.A.); (M.B.); (F.W.S.)
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA; (M.A.); (M.B.); (F.W.S.)
| |
Collapse
|
6
|
Sun LL, Liu Z, Ran F, Huang D, Zhang M, Li XQ, Li WD. Non-coding RNAs regulating endothelial progenitor cells for venous thrombosis: promising therapy and innovation. Stem Cell Res Ther 2024; 15:7. [PMID: 38169418 PMCID: PMC10762949 DOI: 10.1186/s13287-023-03621-z] [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: 09/19/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Venous thromboembolism, which includes deep venous thrombosis (DVT) and pulmonary embolism, is the third most common vascular disease in the world and seriously threatens the lives of patients. Currently, the effect of conventional treatments on DVT is limited. Endothelial progenitor cells (EPCs) play an important role in the resolution and recanalization of DVT, but an unfavorable microenvironment reduces EPC function. Non-coding RNAs, especially long non-coding RNAs and microRNAs, play a crucial role in improving the biological function of EPCs. Non-coding RNAs have become clinical biomarkers of diseases and are expected to serve as new targets for disease intervention. A theoretical and experimental basis for the development of new methods for preventing and treating DVT in the clinic will be provided by studies on the role and molecular mechanism of non-coding RNAs regulating EPC function in the occurrence and development of DVT. To summarize, the characteristics of venous thrombosis, the regulatory role of EPCs in venous thrombosis, and the effect of non-coding RNAs regulating EPCs on venous thrombosis are reviewed. This summary serves as a useful reference and theoretical basis for research into the diagnosis, prevention, treatment, and prognosis of venous thrombosis.
Collapse
Affiliation(s)
- Li-Li Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Zhao Liu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Feng Ran
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Dian Huang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Ming Zhang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Wen-Dong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliate Hospital of Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
7
|
Amirlatifi S, Kooshari Z, Salmani K, Fallah Ziyarani M, Azizi S, Ghotbi E, Zolali B. Evaluation of long noncoding RNA (LncRNA) in pathogenesis of HELLP syndrome: diagnostic and future approach. J OBSTET GYNAECOL 2023; 43:2174836. [PMID: 36795605 DOI: 10.1080/01443615.2023.2174836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
HELLP syndrome is a disorder during pregnancy which is defined by elevation of liver enzymes, haemolysis, and low platelet count. This syndrome is a multifactorial one and both genetic and environmental components can have a crucial role in this syndrome's pathogenesis. Long noncoding RNAs (lncRNAs), are defined as long non-protein coding molecules (more than 200 nucleotides), which are functional units in most cellular processes such as cell cycle, differentiation, metabolism and some diseases progression. As these markers discovered, there has been some evidence that they have an important role in the function of some organs, such as placenta; therefore, alteration and dysregulation of these RNAs can develop or alleviate HELLP disorder. Although the role of lncRNAs has been shown in HELLP syndrome, the process is still unclear. In this review, our purpose is to evaluate the association between molecular mechanisms of lncRNAs and HELLP syndrome pathogenicity to elicit some novel approaches for HELLP diagnosis and treatment.
Collapse
Affiliation(s)
- Shahrzad Amirlatifi
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Kooshari
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kiana Salmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Fallah Ziyarani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Azizi
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elena Ghotbi
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Bita Zolali
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Wang J, Zhan H, Wang M, Song H, Sun J, Zhao G. Sonic hedgehog signaling promotes angiogenesis of endothelial progenitor cells to improve pressure ulcers healing by PI3K/AKT/eNOS signaling. Aging (Albany NY) 2023; 15:10540-10548. [PMID: 37815888 PMCID: PMC10599757 DOI: 10.18632/aging.205093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/21/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Pressure ulcer is a severe disease in the paralyzed and aging populations. Endothelial progenitor cells (EPCs) are able to regulate ulcer healing by modulating angiogenesis, but the molecular mechanism is still obscure. Sonic hedgehog (SHH) signaling contributes to angiogenesis in various diseases and has been identified to modulate EPCs function. Here, we aimed to explore the significance of SHH signaling in EPCs function during pressure ulcers. METHODS The EPCs were isolated and characterized by the expression of DiI-acLDL and bind fluorescein iso-thiocyanate UEA-1. Cell proliferation was detected by cell counting kit 8 (CCK-8). The DiI-acLDL and bind fluorescein iso-thiocyanate UEA-1 were analyzed by immunofluorescent analysis. The angiogenesis of EPCs was analyzed by tube formation assay. The pressure ulcers rat model was constructed, the wound injury was analyzed by H&E staining and angiogenesis was analyzed by the accumulation of CD31 based on immunofluorescent analysis. RESULTS The expression of patched-1 and Gli-1 was enhanced by SHH activator SAG but reduced by SHH inhibitor cyclopamine in the EPCsThe PI3K, Akt, eNOS expression and the Akt phosphorylation were induced by SAG, while the treatment of cyclopamine presented a reversed result. The proliferation and migration of EPCs were enhanced by SAG but repressed by cyclopamine or PI3K/AKT/eNOS signaling inhibitor Y294002, in which the co-treatment of Y294002 could reverse the effect of SAG. CONCLUSIONS Thus, we found that SHH signaling activated angiogenesis properties of EPCs to improve pressure ulcers healing by PI3K/AKT/eNOS signaling. SHH signaling may serve as the potential target for attenuating pressure ulcers.
Collapse
Affiliation(s)
- Jianhua Wang
- Department of Orthopaedics, Jinan Central Hospital, Jinan, Shandong Province, China
| | - Hongyan Zhan
- Department of B-Ultrasound, Fourth People’s Hospital of Jinan, Jinan, Shandong Province, China
| | - Mingming Wang
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong Province, China
| | - Hua Song
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong Province, China
| | - Jianhua Sun
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong Province, China
| | - Gang Zhao
- Department of Orthopaedics, Jinan Central Hospital, Jinan, Shandong Province, China
| |
Collapse
|
9
|
Duan SL, Fu WJ, Jiang YK, Peng LS, Ousmane D, Zhang ZJ, Wang JP. Emerging role of exosome-derived non-coding RNAs in tumor-associated angiogenesis of tumor microenvironment. Front Mol Biosci 2023; 10:1220193. [PMID: 37602326 PMCID: PMC10436220 DOI: 10.3389/fmolb.2023.1220193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
The tumor microenvironment (TME) is an intricate ecosystem that is actively involved in various stages of cancer occurrence and development. Some characteristics of tumor biological behavior, such as proliferation, migration, invasion, inhibition of apoptosis, immune escape, angiogenesis, and metabolic reprogramming, are affected by TME. Studies have shown that non-coding RNAs, especially long-chain non-coding RNAs and microRNAs in cancer-derived exosomes, facilitate intercellular communication as a mechanism for regulating angiogenesis. They stimulate tumor growth, as well as angiogenesis, metastasis, and reprogramming of the TME. Exploring the relationship between exogenous non-coding RNAs and tumor-associated endothelial cells, as well as their role in angiogenesis, clinicians will gain new insights into treatment as a result.
Collapse
Affiliation(s)
- Sai-Li Duan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei-Jie Fu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ying-Ke Jiang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Lu-Shan Peng
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha, China
| | - Diabate Ousmane
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha, China
| | - Zhe-Jia Zhang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jun-Pu Wang
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Bhattacharjee R, Prabhakar N, Kumar L, Bhattacharjee A, Kar S, Malik S, Kumar D, Ruokolainen J, Negi A, Jha NK, Kesari KK. Crosstalk between long noncoding RNA and microRNA in Cancer. Cell Oncol (Dordr) 2023; 46:885-908. [PMID: 37245177 PMCID: PMC10356678 DOI: 10.1007/s13402-023-00806-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2023] [Indexed: 05/29/2023] Open
Abstract
miRNAs and lncRNAs play a central role in cancer-associated gene regulations. The dysregulated expression of lncRNAs has been reported as a hallmark of cancer progression, acting as an independent prediction marker for an individual cancer patient. The interplay of miRNA and lncRNA decides the variation of tumorigenesis that could be mediated by acting as sponges for endogenous RNAs, regulating miRNA decay, mediating intra-chromosomal interactions, and modulating epigenetic components. This paper focuses on the influence of crosstalk between lncRNA and miRNA on cancer hallmarks such as epithelial-mesenchymal transition, hijacking cell death, metastasis, and invasion. Other cellular roles of crosstalks, such as neovascularization, vascular mimicry, and angiogenesis were also discussed. Additionally, we reviewed crosstalk mechanism with specific host immune responses and targeting interplay (between lncRNA and miRNA) in cancer diagnosis and management.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Neeraj Prabhakar
- Centre for Structural System Biology, Department of Physics, University of Hamburg, c/o DESY, Building 15, Notkestr. 852267, Hamburg, Germany
- Pharmacy, Abo Akademi University, Tykistökatu 6A, Turku, Finland
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India
| | - Arkadyuti Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, 834001, India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, Espoo, 00076, Finland
| | - Arvind Negi
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, 00076, Finland.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, 201310, UP, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo, 00076, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, Biocentre 3, Helsinki, Finland.
| |
Collapse
|
11
|
Shao W, Li Z, Wang B, Gong S, Wang P, Song B, Chen Z, Feng Y. Dimethyloxalylglycine Attenuates Steroid-Associated Endothelial Progenitor Cell Impairment and Osteonecrosis of the Femoral Head by Regulating the HIF-1α Signaling Pathway. Biomedicines 2023; 11:biomedicines11040992. [PMID: 37189610 DOI: 10.3390/biomedicines11040992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 05/17/2023] Open
Abstract
Endothelial impairment and dysfunction are closely related to the pathogenesis of steroid-associated osteonecrosis of the femoral head (SONFH). Recent studies have showed that hypoxia inducible factor-1α (HIF-1α) plays a crucial role in endothelial homeostasis maintenance. Dimethyloxalylglycine (DMOG) could suppress HIF-1 degradation and result in nucleus stabilization by repressing prolyl hydroxylase domain (PHD) enzymatic activity. Our results showed that methylprednisolone (MPS) remarkably undermined biological function of endothelial progenitor cells (EPC) by inhibiting colony formation, migration, angiogenesis, and stimulating senescence of EPCs, while DMOG treatment alleviated these effects by promoting HIF-1α signaling pathway, as evidenced by senescence-associated β-galactosidase (SA-β-Gal) staining, colony-forming unit, matrigel tube formation, and transwell assays. The levels of proteins related to angiogenesis were determined by ELISA and Western blotting. In addition, active HIF-1α bolstered the targeting and homing of endogenous EPCs to the injured endothelium in the femoral head. Histopathologically, our in vivo study showed that DMOG not only alleviated glucocorticoid-induced osteonecrosis but also promoted angiogenesis and osteogenesis in the femoral head as detected by microcomputed tomography (Micro-CT) analysis and histological staining of OCN, TRAP, and Factor Ⅷ. However, all of these effects were impaired by an HIF-1α inhibitor. These findings demonstrate that targeting HIF-1α in EPCs may constitute a novel therapeutic approach for the treatment of SONFH.
Collapse
Affiliation(s)
- Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Song Gong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Beite Song
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixiang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
12
|
Zhang B, Qin J. LINC00659 exacerbates endothelial progenitor cell dysfunction in deep vein thrombosis of the lower extremities by activating DNMT3A-mediated FGF1 promoter methylation. Thromb J 2023; 21:24. [PMID: 36890543 PMCID: PMC9996960 DOI: 10.1186/s12959-023-00462-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 03/10/2023] Open
Abstract
It has been shown that long non-coding RNA (lncRNA) LINC00659 was markedly upregulated in the peripheral blood of patients with deep venous thrombosis (DVT). However, the function of LINC00659 in lower extremity DVT (LEDVT) remains to be largely unrevealed. A total of 30 inferior vena cava (IVC) tissue samples and peripheral blood (60 ml per subject) were obtained from LEDVT patients (n = 15) and healthy donors (n = 15), and then LINC00659 expression was detected by RT-qPCR. The results displayed that LINC00659 is upregulated in IVC tissues and isolated endothelial group cells (EPCs) of patients with LEDVT. LINC00659 knock-down promoted the proliferation, migration, and angiogenesis ability of EPCs, while an pcDNA-eukaryotic translation initiation factor 4A3 (EIF4A3), a EIF4A3 overexpression vector, or fibroblast growth factor 1 (FGF1) small interfering RNA (siRNA) combined with LINC00659 siRNA could not enhance this effect. Mechanistically, LINC00659 bound with EIF4A3 promoter to upregulated EIF4A3 expression. Besides, EIF4A3 could facilitate FGF1 methylation and its downregulated expression by recruiting DNA methyltransferases 3A (DNMT3A) to the FGF1 promoter region. Additionally, LINC00659 inhibition could alleviate LEDVT in mice. In summary, the data indicated the roles of LINC00659 in the pathogenesis of LEDVT, and the LINC00659/EIF4A3/FGF1 axis could be a novel therapeutic target for the treatment of LEDVT.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Peripheral Vessel, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China.
| | - Jie Qin
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710061, Shanxi, China
| |
Collapse
|
13
|
Yang Y, Zheng W, Tan W, Wu X, Dai Z, Li Z, Yan Z, Ji Y, Wang Y, Su W, Zhong S, Li Y, Sun Y, Li S, Huang W. Injectable MMP1-sensitive microspheres with spatiotemporally controlled exosome release promote neovascularized bone healing. Acta Biomater 2023; 157:321-336. [PMID: 36481504 DOI: 10.1016/j.actbio.2022.11.065] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Bone marrow mesenchymal stromal cell-derived exosomes (BMSC-Exos) can recruit stem cells for bone repair, with neovessels serving as the main migratory channel for stem cells to the injury site. However, existing exosome (Exo) delivery strategies cannot reach the angiogenesis phase following bone injury. To that end, an enzyme-sensitive Exo delivery material that responds to neovessel formation during the angiogenesis phase was designed in the present study to achieve spatiotemporally controlled Exo release. Herein, matrix metalloproteinase-1 (MMP1) was found to be highly expressed in neovascularized bone; as a result, we proposed an injectable MMP1-sensitive hydrogel microspheres (KGE) made using a microfluidic chip prepared by mixing self-assembling peptide (KLDL-MMP1), GelMA, and BMSC-Exos. The results revealed that KGE microspheres had a uniform diameter of 50-70 µm, ideal for minimally invasive injection and could release exosomes in response to MMP1 expression. In vitro experiments demonstrated that KGE had less cytotoxicity and could promote the migration and osteodifferentiation of BMSCs. Furthermore, in vivo experiments confirmed that KGE could promote bone repair during angiogenesis by recruiting CD90+ stem cells via neovessels. Collectively, our results suggest that injectable enzyme-responsive KGE microspheres could be a promising Exo-secreting material for accelerating neovascularized bone healing. STATEMENT OF SIGNIFICANCE: Exosomes can spread through blood vessels and activate stem cells to participate in bone repair, but under normal circumstances, exosomes lacking sustained-release delivery materials cannot be maintained until the angiogenesis phase. In this study, we found that MMP1 was highly expressed in neovascularized bone, then we proposed an MMP1-sensitive injectable microsphere that carries exosomes and responds temporally and spatially to neovascularization, which maximizes the ability of exosomes to recruit stem cells. Different from previous strategies that focus on promoting angiogenesis to accelerate bone healing, this is a brand new delivery strategy that is stimuli-responsive to neovessel formation. In addition, the preparation of self-assembled peptide microspheres by a microfluidic chip is also proposed for the first time.
Collapse
Affiliation(s)
- Yang Yang
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weihan Zheng
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Tan
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
| | - Xiaoqi Wu
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Zhenning Dai
- Department of Stomatology, Guangdong Key Laboratory of Traditional Chinese Medicine Research and Development, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou 510095, China
| | - Ziyue Li
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zi Yan
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuelun Ji
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China
| | - Yilin Wang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weiwei Su
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shu Zhong
- Department of orthopedic, Dongguan People's Hospital, Dongguan 523058, China
| | - Yanbing Li
- Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Sun
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510630, China.
| | - Shiyu Li
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Wenhua Huang
- The Third Affiliated Hospital of Southern Medical University, Guangdong Medical Innovation Platform for Translation of 3D Printing Application, Southern Medical University, Guangzhou 510630, China; Department of Anatomy, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
14
|
Zhang D, Cui Y, Zhao M, Zheng X, Li C, Wei J, Wang K, Cui J. Orexin-A exerts neuroprotective effect in experimental intracerebral hemorrhage by suppressing autophagy via OXR1-mediated ERK/mTOR signaling pathway. Front Cell Neurosci 2022; 16:1045034. [PMID: 36619670 PMCID: PMC9815810 DOI: 10.3389/fncel.2022.1045034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background Orexin-A (OXA) is a polypeptide produced in the hypothalamus, which binds to specific receptors and exerts multiple physiological effects. Autophagy plays a vital role in early brain injury (EBI) after intracerebral hemorrhage (ICH). However, the relationship between OXA and autophagy after ICH has not been confirmed. Methods In this study, the protective role of OXA was investigated in a model of hemin-induced injury in PC12 cells and blood-injection ICH model in rats, and its potential molecular mechanism was clarified. Neurobehavioral tests, brain water content, and pathologic morphology were assessed after ICH. Cell survival rate was determined using Cell Counting Kit-8 (CCK-8), while apoptosis was detected using flow cytometry. The autophagy protein LC3 that was originally identified as microtubule-associated protein 1 light 3 was evaluated by immunohistochemistry. The ultrastructural changes of cells following ICH were observed by transmission electron microscopy. Western blotting was performed to determine the expression levels of LC3, p62/SQSTM1 (p62), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2), total extracellular signal-regulated kinase 1/2 (t-ERK1/2), mammalian target of rapamycin (mTOR), and phosphorylated mammalian target of rapamycin (p-mTOR). Results OXA treatment significantly improved neurofunctional outcomes, reduced brain edema, and alleviated neuronal apoptosis. OXA administration upregulated p-mTOR and p62, while it downregulated p-ERK1/2 and LC3; this effect was reversed by the orexin receptor 1 (OXR1) antagonist SB-334867. Conclusions This study demonstrates that OXA suppresses autophagy via the OXR1-mediated ERK/mTOR signaling pathway to exert neuroprotective effects, and it might provide a novel therapeutic approach in patients suffering from ICH.
Collapse
Affiliation(s)
- Dexin Zhang
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Ying Cui
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, China
| | - Manman Zhao
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Xuecheng Zheng
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Chunyan Li
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingbo Wei
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Kaijie Wang
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Jianzhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, China,Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China,*Correspondence: Jianzhong Cui,
| |
Collapse
|
15
|
Singh D, Rai V, Agrawal DK. Non-Coding RNAs in Regulating Plaque Progression and Remodeling of Extracellular Matrix in Atherosclerosis. Int J Mol Sci 2022; 23:13731. [PMID: 36430208 PMCID: PMC9692922 DOI: 10.3390/ijms232213731] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022] Open
Abstract
Non-coding RNAs (ncRNAs) regulate cell proliferation, migration, differentiation, inflammation, metabolism of clinically important biomolecules, and other cellular processes. They do not encode proteins but are involved in the regulatory network of various proteins that are directly related to the pathogenesis of diseases. Little is known about the ncRNA-associated mechanisms of atherosclerosis and related cardiovascular disorders. Remodeling of the extracellular matrix (ECM) is critical in the pathogenesis of atherosclerosis and related disorders; however, its regulatory proteins are the potential subjects to explore with special emphasis on epigenetic regulatory components. The activity of regulatory proteins involved in ECM remodeling is regulated by various ncRNA molecules, as evident from recent research. Thus, it is important to critically evaluate the existing literature to enhance the understanding of nc-RNAs-regulated molecular mechanisms regulating ECM components, remodeling, and progression of atherosclerosis. This is crucial since deregulated ECM remodeling contributes to atherosclerosis. Thus, an in-depth understanding of ncRNA-associated ECM remodeling may identify novel targets for the treatment of atherosclerosis and other cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
16
|
Li Y, Jiang X, Zhang Z, Liu J, Wu C, Chen Y, Zhou J, Zhang J, Zhang X. Autophagy promotes directed migration of HUVEC in response to electric fields through the ROS/SIRT1/FOXO1 pathway. Free Radic Biol Med 2022; 192:213-223. [PMID: 36162742 DOI: 10.1016/j.freeradbiomed.2022.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 10/31/2022]
Abstract
Endogenous electric fields (EFs) have been confirmed to facilitate angiogenesis through guiding directional migration of endothelial cells (ECs), but the underlying mechanisms remain obscure. Recent studies suggest that the directed migration of ECs in angiogenesis is correlated with autophagy, and the latter of which could be augmented by EFs. We hypothesize that autophagy may participate in the EFs-guided migration of ECs during angiogenesis. Herein, we showed that EFs induced human umbilical vein endothelial cells (HUVEC) migration toward the cathode with enhanced autophagy. Genetic ablation of autophagy by silencing the autophagy-related gene (Atg) 5 abolished the EFs-directed migration of HUVEC, indicating that autophagy is definitely required for EFs-guided migration of cells. Mechanistically, we identified the intracellular reactive oxygen species (ROS) as a crucial mediator in EFs-triggered autophagy through augmenting the silencing information regulator 2 related enzyme1 (SIRT1)/forkhead box protein O1 (FOXO1) signaling. Either ROS scavenging or SIRT1 knockdown eliminated the EFs-triggered autophagy in HUVEC. Further study showed that SIRT1 promoted FOXO1 deacetylation, facilitating its nuclear accumulation and transcriptional activity, and thereby activating autophagy in EFs-treated HUVECs. In conclusion, our study demonstrated a pivotal role for autophagy in EFs-induced directed migration of HUVECs through the ROS/SIRT1/FOXO1 pathway, and provided a novel theoretical foundation for angiogenesis.
Collapse
Affiliation(s)
- Yi Li
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Xupin Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ze Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jie Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chao Wu
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ying Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Junli Zhou
- Department of Plastic Aesthetic and Burns Surgery, First Affiliated Hospital of Xiamen University and the Teaching Hospital of Fujian Medical University, Xiamen, Fujian, 361003, China.
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xuanfen Zhang
- Department of Plastic Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
17
|
Mechanical Stretch Induced Skin Regeneration: Molecular and Cellular Mechanism in Skin Soft Tissue Expansion. Int J Mol Sci 2022; 23:ijms23179622. [PMID: 36077018 PMCID: PMC9455829 DOI: 10.3390/ijms23179622] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Skin soft tissue expansion is one of the most basic and commonly used techniques in plastic surgery to obtain excess skin for a variety of medical uses. However, skin soft tissue expansion is faced with many problems, such as long treatment process, poor skin quality, high retraction rate, and complications. Therefore, a deeper understanding of the mechanisms of skin soft tissue expansion is needed. The key to skin soft tissue expansion lies in the mechanical stretch applied to the skin by an inflatable expander. Mechanical stimulation activates multiple signaling pathways through cellular adhesion molecules and regulates gene expression profiles in cells. Meanwhile, various types of cells contribute to skin expansion, including keratinocytes, dermal fibroblasts, and mesenchymal stem cells, which are also regulated by mechanical stretch. This article reviews the molecular and cellular mechanisms of skin regeneration induced by mechanical stretch during skin soft tissue expansion.
Collapse
|
18
|
Hu X, Lei X, Guo J, Fu W, Sun W, Lu Q, Su W, Xu Q, Tu K. The Emerging Role of RNA N6-Methyladenosine Modification in Pancreatic Cancer. Front Oncol 2022; 12:927640. [PMID: 35936737 PMCID: PMC9354683 DOI: 10.3389/fonc.2022.927640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/15/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most common malignant cancers, ranking the seventh highest causes of cancer-related deaths globally. Recently, RNA N6-methyladenosine (m6A) is emerging as one of the most abundant RNA modifications in eukaryote cells, involved in multiple RNA processes including RNA translocation, alternative splicing, maturation, stability, and degradation. As reported, m6A was dynamically and reversibly regulated by its “writers”, “erasers”, and “readers”, Increasing evidence has revealed the vital role of m6A modification in the development of multiple types of cancers including PC. Currently, aberrant m6A modification level has been found in both PC tissues and cell lines. Moreover, abnormal expressions of m6A regulators and m6A-modified genes have been reported to contribute to the malignant development of PC. Here in this review, we will focus on the function and molecular mechanism of m6A-modulated RNAs including coding RNAs as well as non-coding RNAs. Then the m6A regulators will be summarized to reveal their potential applications in the clinical diagnosis, prognosis, and therapeutics of PC.
Collapse
Affiliation(s)
- Xiaoge Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiangxiang Lei
- Institute of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wen Fu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wen Sun
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiliang Lu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Wei Su
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Innovation Center for the Study of Pancreatic Diseases, Hangzhou, China
- *Correspondence: Wei Su, ; Qiuran Xu, ; Kangsheng Tu,
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Wei Su, ; Qiuran Xu, ; Kangsheng Tu,
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wei Su, ; Qiuran Xu, ; Kangsheng Tu,
| |
Collapse
|
19
|
Oxidative stress-induced endothelial cells-derived exosomes accelerate skin flap survival through Lnc NEAT1-mediated promotion of endothelial progenitor cell function. Stem Cell Res Ther 2022; 13:325. [PMID: 35850692 PMCID: PMC9290268 DOI: 10.1186/s13287-022-03013-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background Flap transplantation is commonly used in reconstructive surgery. A prerequisite for skin flap survival is sufficient blood supply. However, such approaches remain unclear. This study aimed to explore the underlying mechanisms of exosomes derived from human umbilical vascular endothelial cells (HUVECs) exposed to oxidative stress on endothelial progenitor cells (EPCs) and their subsequent influence on the survival of skin flaps. Methods HUVECs were treated with various concentrations of H2O2 to establish an oxidative stress model. To investigate the effects of H2O2-HUVEC-Exos and HUVEC-Exos, Cell Counting Kit-8, tube formation, invasion assays, and quantitative real-time polymerase chain reaction (qRT-PCR) were performed in EPCs. Microarray analysis was used to reveal the differentially expressed long non-coding RNAs (lncRNAs) in the H2O2-HUVEC-Exos and HUVEC-Exos. In addition, gene silencing and western blotting were employed to determine the mechanism behind lncRNA nuclear enrichment enriched transcript 1 (Lnc NEAT1) in EPCs. Further, a rat skin flap model was used to determine the role of the exosomes in skin flap survival in vivo. Results HUVECs were stimulated with 100 μmol/L H2O2 for 12 h to establish an oxidative stress model. H2O2-HUVEC-Exos promoted the proliferation, tube formation, and invasion of EPCs and remarkably increased skin flap survival compared to the HUVEC-Exos and control groups. Sequencing of exosome RNAs revealed that the Lnc NEAT1 level was dramatically increased in the H2O2-HUVEC-Exos, leading to activation of the Wnt/β-catenin signaling pathway. Comparatively, knockdown of Lnc NEAT1 in HUVEC-Exos and H2O2-HUVEC-Exos significantly inhibits the angiogenic capacity of EPCs, reduced the survival area of skin flap and downregulated the expression levels of Wnt/β-catenin signaling pathway proteins, whereas Wnt agonist partly reversed the negative effect of NEAT1 downregulation on EPCs through the Wnt/β-catenin signaling pathway. Conclusions Exosomes derived from HUVECs stimulated by oxidative stress significantly promoted the pro-angiogenic ability of EPCs through the Wnt/β-catenin signaling pathway mediated by Lnc NEAT1 and hence enhanced random flap survival in vivo. Therefore, the application of H2O2-HUVEC-Exos may serve as an alternative therapy for improving random skin flap survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03013-9.
Collapse
|
20
|
Li C, Pan Y, Tan Y, Wang Y, Sun X. PINK1-Dependent Mitophagy Reduced Endothelial Hyperpermeability and Cell Migration Capacity Under Simulated Microgravity. Front Cell Dev Biol 2022; 10:896014. [PMID: 35874841 PMCID: PMC9300855 DOI: 10.3389/fcell.2022.896014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
The effect of cardiovascular dysfunction including orthostatic intolerance and disability on physical exercise is one of the health problems induced by long-term spaceflight astronauts face. As an important part of vascular structure, the vascular endothelium, uniquely sensitive to mechanical force, plays a pivotal role in coordinating vascular functions. Our study found that simulated microgravity induced PINK1-dependent mitophagy in human umbilical vein endothelial cells (HUVECs). Here, we explored the underlying mechanism of mitophagy induction. The ER stress induced by proteostasis failure in HUVECs promoted the Ca2+ transfer from ER to mitochondria, resulting in mitochondria Ca2+ overload, decreased mitochondrial membrane potential, mitochondria fission, and accumulation of Parkin and p62 in mitochondria and mitophagy under simulated microgravity. Moreover, we assumed that mitophagy played a vital role in functional changes in endothelial cells under simulated microgravity. Using mdivi-1 and PINK1 knockdown, we found that NLRP3 inflammasome activation was enhanced after mitophagy was inhibited. The NLRP3 inflammasome contributed to endothelial hyperpermeability and cellular migration by releasing IL-1β. Thus, mitophagy inhibited cell migration ability and hyperpermeability in HUVECs exposed to clinostat-simulated microgravity. Collectively, we here clarify the mechanism of mitophagy induction by simulated microgravity in vitro and demonstrate the relationship between mitophagy and vascular endothelial functional changes including cellular migration and permeability. This study deepens the understanding of vascular functional changes under microgravity.
Collapse
Affiliation(s)
- Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yingjun Tan
- China Astronaut Research and Training Center, Beijing, China
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| |
Collapse
|
21
|
Li S, Han Y, Lu M, Liu Z, Jin J, Guo Q, Wang Y, Liu H. Mesenchymal stem cell‐exosome‐mediated matrix metalloproteinase 1 participates in oral leukoplakia and carcinogenesis by inducing angiogenesis. J Oral Pathol Med 2022; 51:638-648. [PMID: 35792829 DOI: 10.1111/jop.13321] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/04/2022] [Accepted: 02/10/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Shufang Li
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| | - Ying Han
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| | - Mingxing Lu
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| | - Zijian Liu
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| | - Jianqiu Jin
- Department of Stomatology Beijing Hospital, National Center of Gerontology Beijing China
| | - Qianyun Guo
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| | - Yixiang Wang
- Department of Central Laboratory Peking University School and Hospital of Stomatology Beijing China
| | - Hongwei Liu
- Department of Oral Medicine Peking University School and Hospital of Stomatology Beijing China
| |
Collapse
|
22
|
Yang B, Zhang Z. Suppression of long intergenic non-protein coding RNA 1123 constrains lower extremity deep vein thrombosis via microRNA-125a-3p to target interleukin 1 receptor type 1. Bioengineered 2022; 13:13452-13461. [PMID: 35659191 PMCID: PMC9275874 DOI: 10.1080/21655979.2022.2076496] [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] [Indexed: 12/05/2022] Open
Abstract
Lower extremity deep vein thrombosis (LEDVT) is a disorder of venous return caused by abnormal blood clotting. LEDVT can obstruct the lumen and is the third most common vascular disease after cerebrovascular disease and coronary artery disease. LncRNAs are associated with thrombosis and potentially affect the pathogenesis of DVT. However, no studies have reported the effect of LINC01123 on LEDVT. The aim of this study was to investigate the effect of LINC01123 on LEDVT in rats via the miR-125a-3p/interleukin 1 receptor type 1 (IL1R1) axis. Lentiviral vectors that altering LINC01123, miR-125a-3p and IL1R1 expression were pre-injected into the tail vein of rats, and an LEDVT model was established 1 day later. Detection of LINC01123, miR-125a-3p and IL1R1 expression was performed. Inflammatory factors in femoral venous blood, the length and weight of the thrombus, the histomorphological changes were determined in the rat model. The targeting relation of miR-125a-3p with LINC01123 or IL1R1 was verified. The results presented that LEDVT rats expressed high LINC01123 and IL1R1 and low miR-125a-3p expression levels. After silencing LINC01123 or elevating miR-125a-3p, the rate of thrombosis, length and weight of thrombus, and levels of inflammatory factors were reduced. The targeting relation was presented between miR-125a-3p with LINC01123 or IL1R1. Elevating IL1R1 was available to turn around the action of silence of LINC01123 on LEDVT rats. All in all, suppression of LINC01123 restrains LEDVT via miR-125a-3p to target IL1R1.
Collapse
Affiliation(s)
- Baocai Yang
- Department of General surgery, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
- Department of Vascular Surgery, Yancheng First People’s Hospital of Jiangsu Province, Yancheng, Jiangsu Province, China
| | - ZiXiang Zhang
- Department of General surgery, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| |
Collapse
|
23
|
Huang H, Huang W. Regulation of Endothelial Progenitor Cell Functions in Ischemic Heart Disease: New Therapeutic Targets for Cardiac Remodeling and Repair. Front Cardiovasc Med 2022; 9:896782. [PMID: 35677696 PMCID: PMC9167961 DOI: 10.3389/fcvm.2022.896782] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022] Open
Abstract
Ischemic heart disease (IHD) is the leading cause of morbidity and mortality worldwide. Ischemia and hypoxia following myocardial infarction (MI) cause subsequent cardiomyocyte (CM) loss, cardiac remodeling, and heart failure. Endothelial progenitor cells (EPCs) are involved in vasculogenesis, angiogenesis and paracrine effects and thus have important clinical value in alternative processes for repairing damaged hearts. In fact, this study showed that the endogenous repair of EPCs may not be limited to a single cell type. EPC interactions with cardiac cell populations and mesenchymal stem cells (MSCs) in ischemic heart disease can attenuate cardiac inflammation and oxidative stress in a microenvironment, regulate cell survival and apoptosis, nourish CMs, enhance mature neovascularization, alleviate adverse ventricular remodeling after infarction and enhance ventricular function. In this review, we introduce the definition and discuss the origin and biological characteristics of EPCs and summarize the mechanisms of EPC recruitment in ischemic heart disease. We focus on the crosstalk between EPCs and endothelial cells (ECs), smooth muscle cells (SMCs), CMs, cardiac fibroblasts (CFs), cardiac progenitor cells (CPCs), and MSCs during cardiac remodeling and repair. Finally, we discuss the translation of EPC therapy to the clinic and treatment strategies.
Collapse
|
24
|
Lin Q, Shi Y, Liu Z, Mehrpour M, Hamaï A, Gong C. Non-coding RNAs as new autophagy regulators in cancer progression. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166293. [PMID: 34688868 DOI: 10.1016/j.bbadis.2021.166293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/17/2021] [Accepted: 10/10/2021] [Indexed: 12/09/2022]
Abstract
Recent advances highlight that non-coding RNAs (ncRNAs) are emerging as fundamental regulators in various physiological as well as pathological processes by regulating macro-autophagy. Studies have disclosed that macro-autophagy, which is a highly conserved process involving cellular nutrients, components, and recycling of organelles, can be either selective or non-selective and ncRNAs show their regulation on selective autophagy as well as non-selective autophagy. The abnormal expression of ncRNAs will result in the impairment of autophagy and contribute to carcinogenesis and cancer progression by regulating both selective autophagy as well as non-selective autophagy. This review focuses on the regulatory roles of ncRNAs in autophagy and their involvement in cancer which may provide valuable therapeutic targets for cancer management.
Collapse
Affiliation(s)
- Qun Lin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Yu Shi
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Zihao Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Bioland Laboratory, 510005 Guangzhou, China
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, 75993, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75993 Paris, France
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, 75993, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75993 Paris, France
| | - Chang Gong
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Bioland Laboratory, 510005 Guangzhou, China.
| |
Collapse
|
25
|
Zhang Y, Xu Y, Zhou K, Kao G, Xiao J. MicroRNA‑126 and VEGF enhance the function of endothelial progenitor cells in acute myocardial infarction. Exp Ther Med 2021; 23:142. [PMID: 35069823 PMCID: PMC8756429 DOI: 10.3892/etm.2021.11065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 10/08/2021] [Indexed: 11/06/2022] Open
Abstract
Previous studies have found that microRNA-126 (miR-126) overexpression can exert beneficial effects on endothelial function and angiogenesis. The role of miR-126 was previously reported to be by directly limiting the activities of negative regulators of the vascular endothelial growth factor (VEGF) pathway, such as PI3K regulation subunit 2 (PIK3R2). The aim of the present study was to investigate the role of the miR-126/PIK3R2/VEGF axis in endothelial progenitor cells (EPCs) under hypoxic conditions. An in vitro hypoxia model in EPCs was established by exposing EPCs to hypoxia (O2/N2/CO2, 1/94/5) for 72 h, before reverse transcription-quantitative PCR (RT-qPCR) and western blot analyzes were used to measure miR-126 and PIK3R2 expression in EPCs. The proliferation, migration and tube-forming ability of the transfected cells were measured using MTT, Transwell and tube formation assays, respectively. miR-126 expression was found to be lower in EPCs in the hypoxia group compared with that in the control group (P<0.01). The expression of PIK3R2, a direct target gene of miR-126, was found to be higher in the hypoxia group compared with that in the control group (P<0.01). miR-126 mimic and VEGF-plasmid co-transfection improved the proliferation, migration, tube-forming ability and restored the phosphorylation of AKT in EPCs under hypoxic conditions (all P<0.01). In addition, the effects of miR-126 mimic on hypoxia-induced EPCs were reversed by PIK3R2-plasmid co-transfection, whilst the effects of VEGF-plasmid were enhanced further by co-transfection with the miR-126 mimic. In conclusion, miR-126 promoted the functions of EPCs under hypoxic conditions by negatively targeting PIK3R2, whilst the combined overexpression of miR-126 and VEGF enhanced these aforementioned effects.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center (Fourth People's Hospital of Chongqing), Chongqing 400014, P.R. China
| | - Yi Xu
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center (Fourth People's Hospital of Chongqing), Chongqing 400014, P.R. China
| | - Ke Zhou
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center (Fourth People's Hospital of Chongqing), Chongqing 400014, P.R. China
| | - Guoying Kao
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center (Fourth People's Hospital of Chongqing), Chongqing 400014, P.R. China
| | - Jun Xiao
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center (Fourth People's Hospital of Chongqing), Chongqing 400014, P.R. China
| |
Collapse
|
26
|
Qiu X, Lin J, Chen Y, Liang B, Li L. Identification of Hub Genes Associated with Abnormal Endothelial Function in Early Coronary Atherosclerosis. Biochem Genet 2021; 60:1189-1204. [PMID: 34800203 DOI: 10.1007/s10528-021-10139-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022]
Abstract
Abnormal coronary endothelial function is an important step in the development of atherosclerosis. Coronary atherosclerosis is one of the main causes of death worldwide. We constructed a co-expression network to identify hub genes associated with abnormal coronary endothelial function in early coronary atherosclerosis. In brief, we used the GSE132651 dataset from the gene expression omnibus database. The top 5000 genes with greatest variances were used for weighted gene co-expression network analysis, and the module most strongly correlated with abnormal coronary endothelial function was chosen as key module. Functional enrichment analysis was performed for genes in the key module, a protein-protein interaction network was constructed to find hub genes, and gene set enrichment analysis (GSEA) was also performed. Genes were classified into 7 modules, with the midnightblue module being the one that was most related to abnormal coronary endothelial function and containing genes enriched in DNA replication, cell cycle, nucleotide excision repair, and Human T-cell leukemia virus 1 infection. We identified nine hub genes (HOXC5, PRND, PADI3, RC3H1, DAPP1, SIT1, DRICH1, GPRIN2, and RHO), which differently expressed in abnormal and normal coronary endothelial function samples. GSEA suggested that samples associated with abnormal coronary endothelial function and highly expressed hub genes were linked with immune, coagulation, hypoxia, and angiogenesis processes. These hub genes, their expression pattern, and pathways may be involved in the development of abnormal coronary endothelial function and promotion of early coronary atherosclerosis.
Collapse
Affiliation(s)
- Xue Qiu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jinyan Lin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yanbing Chen
- The First Clinical Medical School, Guangxi Medical University, Nanning, 530021, Guangxi Province, People's Republic of China
| | - Bixiao Liang
- The First Clinical Medical School, Guangxi Medical University, Nanning, 530021, Guangxi Province, People's Republic of China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
27
|
Shang B, Xu T, Hu N, Mao Y, Du X. Circ-Klhl8 overexpression increased the therapeutic effect of EPCs in diabetic wound healing via the miR-212-3p/SIRT5 axis. J Diabetes Complications 2021; 35:108020. [PMID: 34507876 DOI: 10.1016/j.jdiacomp.2021.108020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/15/2022]
Abstract
Previous studies found that hypoxic pretreatment of endothelial progenitor cells (EPCs) prior to transplantation had a greater therapeutic effect than untreated EPCs in promoting diabetic wound healing. However, the exact mechanism is uncertain. Here, circRNA expression in EPCs after hypoxic treatment was investigated. High-throughput sequencing was used to assess abnormal expression by EPCs of circular RNAs (circRNAs) following hypoxic pretreatment. Additionally, an in vivo full-thickness skin defect mouse model was used to assess the effects of transplanted EPCs on diabetic wound closure. Subsequently, the regulatory mechanism and targets were studied. The results showed that circ-Klhl8 overexpression suppressed hyper glucose-induced endothelial cell damage by activating autophagy. MiR-212-3p and SIRT5 were identified as the downstream targets of circ-Klhl8. Circ-Klhl8 overexpression promoted skin wound healing by regulating SIRT5-mediated autophagy. In conclusion, the study found that circ-Klhl8 overexpression increased the EPC therapeutic effect in promoting diabetic wound healing by targeting the miR-212-3p/SIRT5 axis.
Collapse
Affiliation(s)
- Bin Shang
- Department of Cardio-Thoracic Surgery, The Second Affiliated Hospital of BBMC, #633 Longhua Road, Huaishang District, Bengbu, 233000 Anhui, China
| | - Tianze Xu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China
| | - Nan Hu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China.
| | - Youjun Mao
- Department of Vascular Surgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, #68 Gehu Zhong Road, Wujin District, Changzhou City 213000, China.
| | - Xiaolong Du
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, #321 Zhongshan Road, Nanjing, 210008 Jiangsu, China.
| |
Collapse
|
28
|
Chen J, Gu S, Song Y, Ji X, Zeng W, Wang X, Wang Y, Feng Q. The impact of cardiomotor rehabilitation on endothelial function in elderly patients with chronic heart failure. BMC Cardiovasc Disord 2021; 21:524. [PMID: 34724901 PMCID: PMC8561974 DOI: 10.1186/s12872-021-02327-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Background To explore the effects of cardiac exercise rehabilitation on peripheral blood endothelial progenitor cells (EPC) in elderly patients with chronic heart failure. Methods 80 elderly patients with chronic heart failure were selected from March 2017 to March 2019 and randomly divided into two groups (N = 40). The control group was treated routinely and walked freely for 30–60 min every day. The patients in the exercise rehabilitation group developed a cardiac exercise rehabilitation plan. Then, cardiac function and peripheral blood B-natriuretic peptide (BNP) levels in the two groups were compared. The cell viability, proliferation, apoptosis, and invasion ability of EPCs were detected. The levels of the PI3K/AKT pathway and eNOS and VEGF were compared. Results There were no significant differences in all indexes between the two groups before treatment (P > 0.05), and both improved significantly after treatment (P < 0.05). After treatment, LVEF and LVFS in the exercise rehabilitation group were significantly higher than those in the control group (P < 0.05), and LVEDD and LVESD were significantly lower than those in the control group (P < 0.05). The BNP level in the exercise rehabilitation group was significantly lower than that in the control group (P < 0.05). The cell viability, proliferation, invasion ability of EPC, and the levels of PI3K, AKT, eNOS, and VEGF mRNA and protein in the exercise rehabilitation group were significantly higher than those in the control group. Apoptosis rate was significantly lower than those in the control group (P < 0.05). Conclusions Visceral exercise rehabilitation can improve cardiac ejection and myocardial function in elderly patients with chronic heart failure, and can promote the vitality, proliferation, and invasion of peripheral blood EPC, and promote the expression of eNOS and VEGF by upregulating the PI3K/AKT pathway to promote angiogenesis and endothelial function. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02327-5.
Collapse
Affiliation(s)
- Juming Chen
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Shenhong Gu
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China.
| | - Yanling Song
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Xinbo Ji
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Wangyuan Zeng
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Xiaoxi Wang
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Yachun Wang
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou City, 570102, Hainan Province, China
| | - Qingfeng Feng
- Graduate School, Hainan Medical University, China Medical University, Haikou City, 571199, Hainan Province, China
| |
Collapse
|
29
|
Wang M, Yang D, Hu Z, Shi Y, Ma Y, Cao X, Guo T, Cai H, Cai H. Extracorporeal Cardiac Shock Waves Therapy Improves the Function of Endothelial Progenitor Cells After Hypoxia Injury via Activating PI3K/Akt/eNOS Signal Pathway. Front Cardiovasc Med 2021; 8:747497. [PMID: 34708093 PMCID: PMC8542843 DOI: 10.3389/fcvm.2021.747497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Extracorporeal cardiac shock waves (ECSW) have great potential in the treatment of coronary heart disease. Endothelial progenitor cells (EPCs) are a class of pluripotent progenitor cells derived from bone marrow or peripheral blood, which have the capacity to migrate to ischemic myocardium and differentiate into mature endothelial cells and play an important role in neovascularization and endothelial repair. In this study, we investigated whether ECSW therapy can improve EPCs dysfunction and apoptosis induced by hypoxia and explored the underlying mechanisms. Methods: EPCs were separated from ApoE gene knockout rat bone marrow and identified using flow cytometry and fluorescence staining. EPCs were used to produce in vitro hypoxia-injury models which were then divided into six groups: Control, Hypoxia, Hypoxia + ECSW, Hypoxia + LY294002 + ECSW, Hypoxia + MK-2206 + ECSW, and Hypoxia + L-NAME + ECSW. EPCs from the Control, Hypoxia, and Hypoxia + ECSW groups were used in mRNA sequencing reactions. mRNA and protein expression levels were analyzed using qRT-PCR and western blot analysis, respectively. Proliferation, apoptosis, adhesion, migration, and angiogenesis were measured using CCK-8, flow cytometry, gelatin, transwell, and tube formation, respectively. Nitric oxide (NO) levels were measured using an NO assay kit. Results: Kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis showed that differentially expressed genes were enriched in cancer signaling, PI3K-Akt signaling, and Rap1 signaling pathways. We selected differentially expressed genes in the PI3K-Akt signaling pathway and verified them using a series of experiments. The results showed that ECSW therapy (500 shots at 0.09 mJ/mm2) significantly improved proliferation, adhesion, migration, and tube formation abilities of EPCs following hypoxic injury, accompanied by upregulation of p-PI3K, p-Akt, p-eNOS, Bcl-2 protein and NO, PI3K, and Akt mRNA expression, and downregulation of Bax and Caspase3 protein expression. All these effects of ECSW were eliminated using inhibitors specific to PI3K (LY294002), Akt (MK-2206), and eNOS (L-NAME). Conclusion: ECSW exerted a strong repaired effect on EPCs suffering inhibited hypoxia injury by inhibiting cell apoptosis and promoting angiogenesis, mainly through activating the PI3K/Akt/eNOS signaling pathway, which provide new evidence for ECSW therapy in CHD.
Collapse
Affiliation(s)
- Mingqiang Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dan Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Hu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingyu Cao
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tao Guo
- Department of Cardiology, Yunnan Fuwai Cardiovascular Hospital, Kunming, China
| | - Hongbo Cai
- Department of Vascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hongyan Cai
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
30
|
Luo L, Yan C, Fuchi N, Kodama Y, Zhang X, Shinji G, Miura K, Sasaki H, Li TS. Mesenchymal stem cell-derived extracellular vesicles as probable triggers of radiation-induced heart disease. Stem Cell Res Ther 2021; 12:422. [PMID: 34294160 PMCID: PMC8296737 DOI: 10.1186/s13287-021-02504-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Radiation-induced heart disease has been reported, but the underlying mechanisms remain unclear. Mesenchymal stem cells (MSCs), also residing in the heart, are highly susceptible to radiation. We examined the hypothesis that the altered secretion of extracellular vesicles (EVs) from MSCs is the trigger of radiation-induced heart disease. METHODS By exposing human placental tissue-derived MSCs to 5 Gy γ-rays, we then isolated EVs from the culture medium 48 h later and evaluated the changes in quantity and quality of EVs from MSCs after radiation exposure. The biological effects of EVs from irradiated MSCs on HUVECs and H9c2 cells were also examined. RESULTS Although the amount and size distribution of EVs did not differ between the nonirradiated and irradiated MSCs, miRNA sequences indicated many upregulated or downregulated miRNAs in irradiated MSCs EVs. In vitro experiments using HUVEC and H9c2 cells showed that irradiated MSC-EVs decreased cell proliferation (P < 0.01), but increased cell apoptosis and DNA damage. Moreover, irradiated MSC-EVs impaired the HUVEC tube formation and induced calcium overload in H9c2 cells. CONCLUSIONS EVs released from irradiated MSCs show altered miRNA profiles and harmful effects on heart cells, which provides new insight into the mechanism of radiation-related heart disease risks.
Collapse
Affiliation(s)
- Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- Medical Technology School of Xuzhou Medical University, Xuzhou Key Laboratory of Laboratory Diagnostics, Tongshan Road 209, Xuzhou, 221004, China
| | - Chen Yan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Naoki Fuchi
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, 852-8523, Japan
| | - Yukinobu Kodama
- Department of Pharmacy, Nagasaki University Hospital, Nagasaki, 852-8523, Japan
| | - Xu Zhang
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Goto Shinji
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Kiyonori Miura
- Department of Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, 852-8523, Japan
| | - Hitoshi Sasaki
- Department of Pharmacy, Nagasaki University Hospital, Nagasaki, 852-8523, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
31
|
Chen C, Gao H, Su X. Autophagy-related signaling pathways are involved in cancer (Review). Exp Ther Med 2021; 22:710. [PMID: 34007319 PMCID: PMC8120650 DOI: 10.3892/etm.2021.10142] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a self-digestion process in cells that can maintain energy homeostasis under normal circumstances. However, misfolded proteins, damaged mitochondria and other unwanted components in cells can be decomposed and reused via autophagy in some specific cases (including hypoxic stress, low energy states or nutrient deprivation). Therefore, autophagy serves a positive role in cell survival and growth. However, excessive autophagy may lead to apoptosis. Furthermore, abnormal autophagy may lead to carcinogenesis and promote tumorigenesis in normal cells. In tumor cells, autophagy may provide the energy required for excessive proliferation, promote the growth of cancer cells, and evade apoptosis caused by certain treatments, including radiotherapy and chemotherapy, resulting in increased treatment resistance and drug resistance. On the other hand, autophagy leads to an insufficient nutrient supply in cancer cells and the destruction of energy homeostasis, thereby inducing cancer cell apoptosis. Therefore, understanding the mechanism of the double-edged sword of autophagy is crucial for the treatment of cancer. The present review summarizes the signaling pathways and key factors involved in autophagy and cancer to provide possible strategies for treating tumors.
Collapse
Affiliation(s)
- Caixia Chen
- Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Hui Gao
- Department of Thoracic Surgery, Inner Mongolia Autonomous Region Cancer Hospital, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xiulan Su
- Clinical Medicine Research Center, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| |
Collapse
|
32
|
Liu Y, Hu R, Zhu J, Nie X, Jiang Y, Hu P, Liu Y, Sun Z. The lncRNA PAHRF functions as a competing endogenous RNA to regulate MST1 expression by sponging miR-23a-3p in pulmonary arterial hypertension. Vascul Pharmacol 2021; 139:106886. [PMID: 34126237 DOI: 10.1016/j.vph.2021.106886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 04/11/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Emerging evidence has shown that long non-coding RNA (lncRNA) plays important roles in the development of pulmonary arterial hypertension (PAH). However, some new lncRNAs in patients with PAH are still lacking research. Herein, we examined the expression and role of lncRNA (pulmonary arterial hypertension related factor, PAHRF) in PAH. METHODS LncRNA PAHRF expression and localization were analyzed by realtime PCR and fluorescence in situ hybridization. Proliferation and apoptosis were detected by MTT, CCK-8, EDU staining, JC-1 assay, flow cytometry and western blotting. Luciferase activity assay was used to identify PAHRF/ miR-23a-3p/serine/threonine kinase 4 (STK4/MST1) interaction. RESULTS LncRNA PAHRF was down-regulated in both the PAs of PAH patients and hypoxic human pulmonary artery smooth muscle cells (PASMCs). The overexpression of PAHRF inhibited the proliferation and promoted the apoptosis of PASMCs. Similarly, we also found PAHRF overexpression decreased the proliferation under hypoxia condition. Knockdown of PAHRF exerted the opposite effects. Luciferase activity assay proved molecular binding between PAHRF and hsa-miR-23a-3p. Moreover, MST1 was confirmed to be the putative target gene and regulated by PAHRF/miR-23a-3p. In addition, we explored the molecular mechanism regulating the expression of miR-23a-3p, and found that lncRNA PAHRF acted as an endogenous sponge for miR-23a-3p, and silencing lncRNA PAHRF could up-regulate the expression of miR-23a-3p. On the contrary, PAHRF-overexpressing plasmid inhibited the expression of miR-23a-3p in hypoxia. CONCLUSIONS Our present study reveals a novel PAH regulating model that is composed of PAHRF, miR-23a-3p, and MST1. The aim of this study is probably going to provide a new explanation and give a further understanding of the occurrence of vascular remodeling in PAH from the perspective competing endogenous RNA hypothesis.
Collapse
Affiliation(s)
- Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang 222061, China.
| | - Rong Hu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang 222061, China
| | - Jinquan Zhu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China
| | - Xiaowei Nie
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen 518112, Guangdong Province, China.; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | - Yanjiao Jiang
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang 222061, China
| | - Panpan Hu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang 222061, China
| | - Yi Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang 222061, China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang 222061, China; Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang 222061, China
| |
Collapse
|
33
|
Zhao Y, Santelli A, Zhu XY, Zhang X, Woollard JR, Chen XJ, Jordan KL, Krier J, Tang H, Saadiq I, Lerman A, Lerman LO. Low-Energy Shockwave Treatment Promotes Endothelial Progenitor Cell Homing to the Stenotic Pig Kidney. Cell Transplant 2021; 29:963689720917342. [PMID: 32237997 PMCID: PMC7444225 DOI: 10.1177/0963689720917342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cells (EPCs) patrols the circulation and contributes to
endothelial cell regeneration. Atherosclerotic renal artery stenosis (ARAS)
induces microvascular loss in the stenotic kidney (STK). Low-energy shockwave
therapy (SW) can induce angiogenesis and restore the STK microcirculation, but
the underlying mechanism remains unclear. We tested the hypothesis that SW
increases EPC homing to the swine STK, associated with capillary regeneration.
Normal pigs and pigs after 3 wk of renal artery stenosis were treated with six
sessions of low-energy SW (biweekly for three consecutive weeks) or left
untreated. Four weeks after completion of treatment, we assessed EPC
(CD34+/KDR+) numbers and levels of the homing-factor stromal cell-derived factor
(SDF)-1 in the inferior vena cava and the STK vein and artery, as well as
urinary levels of vascular endothelial growth factor (VEGF) and integrin-1β.
Subsequently, we assessed STK morphology, capillary count, and expression of the
proangiogenic growth factors angiopoietin-1, VEGF, and endothelial nitric oxide
synthase ex vivo. A 3-wk low-energy SW regimen improved STK
structure, capillary count, and function in ARAS+SW, and EPC numbers and
gradients across the STK decreased. Plasma SDF-1 and renal expression of
angiogenic factors were increased in ARAS+SW, and urinary levels of VEGF and
integrin-1β tended to rise during the SW regimen. In conclusion, SW improves
ischemic kidney capillary density, which is associated with, and may be at least
in part mediated by, promoting EPCs mobilization and homing to the stenotic
kidney.
Collapse
Affiliation(s)
- Yu Zhao
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Institute of Nephrology, Zhong Da Hospital, Southeast University, School of Medicine, Nanjing, Jiangsu, China.,* Both the authors contributed equally to this article
| | - Adrian Santelli
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,* Both the authors contributed equally to this article
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Xin Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Xiao-Jun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - James Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ishran Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Jin L, Hong N, Ai X, Wang J, Li Z, Han Z, Zhang Q, Yu Y, Sun K. LncRNAs as Therapeutic Targets for Autophagy-involved Cardiovascular Diseases: A Review of Molecular Mechanism and T herapy Strategy. Curr Med Chem 2021; 28:1796-1814. [PMID: 32196441 DOI: 10.2174/0929867327666200320161835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/12/2020] [Accepted: 03/06/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. The concept of precision medicine in CVD therapy today requires the incorporation of individual genetic and environmental variability to achieve personalized disease prevention and tailored treatment. Autophagy, an evolutionarily conserved intracellular degradation process, has been demonstrated to be essential in the pathogenesis of various CVDs. Nonetheless, there have been no effective treatments for autophagy- involved CVDs. Long noncoding RNAs (lncRNAs) are noncoding RNA sequences that play versatile roles in autophagy regulation, but much needs to be explored about the relationship between lncRNAs and autophagy-involved CVDs. SUMMARY Increasing evidence has shown that lncRNAs contribute considerably to modulate autophagy in the context of CVDs. In this review, we first summarize the current knowledge of the role lncRNAs play in cardiovascular autophagy and autophagy-involved CVDs. Then, recent developments of antisense oligonucleotides (ASOs) designed to target lncRNAs to specifically modulate autophagy in diseased hearts and vessels are discussed, focusing primarily on structure-activity relationships of distinct chemical modifications and relevant clinical trials. PERSPECTIVE ASOs are promising in cardiovascular drug innovation. We hope that future studies of lncRNA-based therapies would overcome existing technical limitations and help people who suffer from autophagy-involved CVDs.
Collapse
Affiliation(s)
- Lihui Jin
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Nanchao Hong
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Zhuoyan Li
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Zhenyuan Han
- Department of Oral Pathology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yu Yu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| |
Collapse
|
35
|
Down-regulation of miR-361-5p promotes the viability, migration and tube formation of endothelial progenitor cells via targeting FGF1. Biosci Rep 2021; 40:226529. [PMID: 32985665 PMCID: PMC7569154 DOI: 10.1042/bsr20200557] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Transplantion of bone marrow-derived endothelial progenitor cells (EPCs) may be a novel treatment for deep venous thrombosis (DVT). The present study probed into the role of microRNA (miR)-361-5p in EPCs and DVT recanalization. EPCs were isolated from male Sprague-Dawley (SD) rats and identified using confocal microscopy and flow cytometry. The viability, migration and tube formation of EPCs were examined using MTT assay, wound-healing assay and tube formation assay, respectively. Target gene and potential binding sites between miR-361-5p and fibroblast growth factor 1 (FGF1) were predicted by StarBase and confirmed by dual-luciferase reporter assay. Relative expressions of miR-361-5p and FGF1 were detected using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. A DVT model in SD rats was established to investigate the role of EPC with miR-361-5p antagomir in DVT by Hematoxylin-Eosin (H&E) staining. EPC was identified as 87.1% positive for cluster of difference (CD)31, 2.17% positive for CD133, 85.6% positive for von Willebrand factor (vWF) and 94.8% positive for vascular endothelial growth factor receptor-2 (VEGFR2). MiR-361-5p antagomir promoted proliferation, migration and tube formation of EPCs and up-regulated FGF1 expression, thereby dissolving thrombus in the vein of DVT rats. FGF1 was the target of miR-361-5p, and overexpressed FGF1 reversed the effects of up-regulating miR-361-5p on suppressing EPCs. Down-regulation of miR-361-5p enhanced thrombus resolution in vivo and promoted EPC viability, migration and angiogenesis in vitro through targeting FGF1. Therefore, miR-361-5p may be a potential therapeutic target for DVT recanalization.
Collapse
|
36
|
Liang J, Zhang X, Xia W, Tong X, Qiu Y, Qiu Y, He J, Yu B, Huang H, Tao J. Promotion of Aerobic Exercise Induced Angiogenesis Is Associated With Decline in Blood Pressure in Hypertension: Result of EXCAVATION-CHN1. Hypertension 2021; 77:1141-1153. [PMID: 33611934 DOI: 10.1161/hypertensionaha.120.16107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jianwen Liang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.).,Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University (J.L., H.H.)
| | - Xiaoyu Zhang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Wenhao Xia
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Xinzhu Tong
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Yanxia Qiu
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Yumin Qiu
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Jiang He
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Bingbo Yu
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.)
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University (J.L., H.H.)
| | - Jun Tao
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University (J.L., X.Z., W.X., X.T., Yanxia Qiu, Yumin Qiu, J.H., B.Y., J.T.).,Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases (J.T.).,Key Laboratory on Assisted Circulation, Ministry of Health, China (J.T.).,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases (J.T.)
| |
Collapse
|
37
|
Arcucci V, Stacker SA, Achen MG. Control of Gene Expression by Exosome-Derived Non-Coding RNAs in Cancer Angiogenesis and Lymphangiogenesis. Biomolecules 2021; 11:249. [PMID: 33572413 PMCID: PMC7916238 DOI: 10.3390/biom11020249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Abstract: Tumour angiogenesis and lymphangiogenesis are hallmarks of cancer and have been associated with tumour progression, tumour metastasis and poor patient prognosis. Many factors regulate angiogenesis and lymphangiogenesis in cancer including non-coding RNAs which are a category of RNAs that do not encode proteins and have important regulatory functions at transcriptional and post-transcriptional levels. Non-coding RNAs can be encapsulated in extracellular vesicles called exosomes which are secreted by tumour cells or other cells in the tumour microenvironment and can then be taken up by the endothelial cells of blood vessels and lymphatic vessels. The "delivery" of these non-coding RNAs to endothelial cells in tumours can facilitate tumour angiogenesis and lymphangiogenesis. Here we review recent findings about exosomal non-coding RNAs, specifically microRNAs and long non-coding RNAs, which regulate tumour angiogenesis and lymphangiogenesis in cancer. We then focus on the potential use of these molecules as cancer biomarkers and opportunities for exploiting ncRNAs for the treatment of cancer.
Collapse
Affiliation(s)
- Valeria Arcucci
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, 305 Grattan St., Melbourne VIC 3000, Australia; (V.A.); (S.A.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville VIC 3010, Australia
| | - Steven A. Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, 305 Grattan St., Melbourne VIC 3000, Australia; (V.A.); (S.A.S.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville VIC 3010, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville VIC 3050, Australia
| | - Marc G. Achen
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, 9 Princes Street, Fitzroy VIC 3065, Australia
- Department of Medicine, St Vincent’s Hospital, University of Melbourne, Fitzroy VIC 3065, Australia
| |
Collapse
|
38
|
Wang X, Li X, Li J, Zhai L, Liu D, Abdurahman A, Zhang Y, Yokota H, Zhang P. Mechanical loading stimulates bone angiogenesis through enhancing type H vessel formation and downregulating exosomal miR-214-3p from bone marrow-derived mesenchymal stem cells. FASEB J 2021; 35:e21150. [PMID: 33161580 PMCID: PMC7748991 DOI: 10.1096/fj.202001080rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Exosomes are important transporters of miRNAs, which play varying roles in the healing of the bone fracture. Angiogenesis is one of such critical events in bone healing, and we previously reported the stimulatory effect of mechanical loading in vessel remodeling. Focusing on type H vessels and exosomal miR-214-3p, this study examined the mechanism of loading-driven angiogenesis. MiRNA sequencing and qRT-PCR revealed that miR-214-3p was increased in the exosomes of the bone-losing ovariectomized (OVX) mice, while it was significantly decreased by knee loading. Furthermore, compared to the OVX group, exosomes, derived from the loading group, promoted the angiogenesis of endothelial cells. In contrast, exosomes, which were transfected with miR-214-3p, decreased the angiogenic potential. Notably, knee loading significantly improved the microvascular volume, type H vessel formation, and bone mineral density and contents, as well as BV/TV, Tb.Th, Tb.N, and Tb.Sp. In cell cultures, the overexpression of miR-214-3p in endothelial cells reduced the tube formation and cell migration. Collectively, this study demonstrates that knee loading promotes angiogenesis by enhancing the formation of type H vessels and downregulating exosomal miR-214-3p.
Collapse
Affiliation(s)
- Xuetong Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lidong Zhai
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Abdusami Abdurahman
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yifan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, IN 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, IN 46202, USA
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
39
|
Orexin-A protects against cerebral ischemia-reperfusion injury by inhibiting excessive autophagy through OX1R-mediated MAPK/ERK/mTOR pathway. Cell Signal 2020; 79:109839. [PMID: 33212156 DOI: 10.1016/j.cellsig.2020.109839] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Orexin A (OXA) is a neuroprotective peptide that exerts protective effects on multiple physiological and pathological processes. Activation of autophagy is linked to the occurrence of cerebral ischemia-reperfusion injury (CIRI); however, its function remains incompletely understood. In this study, OXA was sought to exert its neuroprotective role by regulating autophagy in oxygen and glucose deprivation and reoxygenation (OGD/R) model and middle cerebral artery occlusion (MCAO) model of rats, and to elucidate the underlying molecular mechanisms. Acridine orange (AO) staining was used to evaluate autophagic vacuoles. Cell viability was measured by CCK8. The levels of p-ERK1/2, t-ERK1/2, p-mTOR, LC3B, Beclin 1, and p62 were evaluated by western blotting. Apoptosis rate was detected by Hoechst 33342 staining and Terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL). OXA treatment alleviated neuronal apoptosis and significantly inhibited autophagy activity. Mechanistically, OXA exerted its neuroprotective effects in vivo and in vitro by suppressing over-activated autophagy by modulating OX1R-mediated MAPK/ERK/mTOR pathway. The results of this study elucidate the roles of autophagy in CIRI and the mechanisms underlying the neuroprotective action of OXA. Our findings could facilitate the development of novel therapeutics for ischemic stroke.
Collapse
|
40
|
Wu Y, Tang Y, Zhang X, Chu Z, Liu Y, Tang C. MMP-1 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via the JNK and ERK pathway. Int J Biochem Cell Biol 2020; 129:105880. [PMID: 33157237 DOI: 10.1016/j.biocel.2020.105880] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/05/2020] [Accepted: 10/12/2020] [Indexed: 01/31/2023]
Abstract
Enhancing the functions of mesenchymal stem cells (MSCs) is considered a potential approach for promoting tissue regeneration. In this study, we investigated the effects of Matrix Metalloproteinase-1 (MMP-1) on bone marrow mesenchymal stem cells (BMSCs) and its mechanism. Our results showed that knockdown of MMP-1 impeded scratch closure, attenuated proliferation, inhibited ALP activity, ALP denser staining and mineralization in vitro, and decreased expression of RUNX2, OSX, OPN and OCN in BMSCs, while 20 ng/mL recombinant human MMP-1 protein (rhMMP-1) significantly accelerated scratch closure, enhanced proliferation, ALP activity, ALP denser staining and mineralization in vitro, and increased expression of RUNX2, OSX, OPN and OCN. In addition, knockdown of MMP-1 inhibited the expression of phosphorylated c-Jun N-terminal kinase (p-JNK) and phosphorylated extracellular regulated protein kinases (p-ERK), while 20 ng/mL rhMMP-1 increased the expression of p-JNK and p-ERK in BMSCs. Furthermore, inhibition of c-Jun N-terminal kinase (JNK) and extracellular regulated protein kinases (ERK) by their inhibitor SP600125 and PD98059 dramatically blocked MMP-1-enhanced ALP activity and mineralization in BMSCs. Our results revealed that MMP-1 could accelerate the osteogenic differentiation potentials of BMSCs via the JNK and ERK pathway, providing the mechanism underlying MSC biology and identifying a potential target for improving bone tissue regeneration.
Collapse
Affiliation(s)
- Yizhen Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yi Tang
- Second Dental Center, School and Hospital of Stomatology, Peking University, Beijing, 100081, PR China
| | - Xiaozhen Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China; Department of Dental Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Zhuangzhuang Chu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yajing Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China
| | - Chunbo Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, PR China; Department of Dental Implantology, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
41
|
Wang M, Xu H, Li Y, Cao C, Zhu H, Wang Y, Zhao Z, Pei G, Zhu F, Yang Q, Deng X, Zhou C, Guo Y, Wu J, Liao W, Yang J, Yao Y, Zeng R. Exogenous bone marrow derived-putative endothelial progenitor cells attenuate ischemia reperfusion-induced vascular injury and renal fibrosis in mice dependent on pericytes. Am J Cancer Res 2020; 10:12144-12157. [PMID: 33204334 PMCID: PMC7667688 DOI: 10.7150/thno.48562] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale: Capillaries are composed of endothelial cells and the surrounding mural cells, pericytes. Microvascular repair after injury involves not only the proliferation of endothelial cells but also pericyte-based vessel stabilization. Exogenous bone marrow derived-putative endothelial progenitor cells (b-pEPCs) have the potential for vascular repair; however, their effect on vascular structure stabilization and pericyte-related pathobiological outcomes in the injured kidney has not been fully examined. Methods: We applied ischemia-reperfusion (IR) to induce renal vascular injury and renal fibrosis in mice. Platelet-derived growth factor receptor β (PDGFR-β)-DTR-positive mice were generated to deplete pericytes, and exogenous b-pEPCs and the PDGFR-β ligand, PDGF chain B (PDGF-BB), were employed to explore the relationship among b-pEPCs, pericytes, vascular repair, and early renal fibrosis. Results: Administration of b-pEPCs reduced IR-induced pericyte-endothelial detachment, pericyte proliferation, and myofibroblast transition via a paracrine mode, which preserved not only vascular stabilization but also ameliorated IR-initiated renal fibrosis. PDGF-BB upregulated the expression of PDGFR-β, exacerbated vascular abnormality, and pericyte-myofibroblast transition, which were ameliorated by b-pEPCs administration. The exogenous b-pEPCs and their culture medium (CM) induced vascular injury protection, and renal fibrosis was blocked by selective deletion of pericytes. Conclusion: Exogenous b-pEPCs directly protect against IR-induced vascular injury and prevent renal fibrosis by inhibiting the activation of PDGFR-β-positive pericytes.
Collapse
|
42
|
Li Y, Zhi K, Han S, Li X, Li M, Lian W, Zhang H, Zhang X. TUG1 enhances high glucose-impaired endothelial progenitor cell function via miR-29c-3p/PDGF-BB/Wnt signaling. Stem Cell Res Ther 2020; 11:441. [PMID: 33059750 PMCID: PMC7558752 DOI: 10.1186/s13287-020-01958-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Diabetes is associated with the dysfunction of endothelial progenitor cells (EPCs), characterized as impaired angiogenesis, a phenomenon thought to be involved in the development of diabetic foot. lncRNA plays an essential role in microvascular dysfunction and signaling pathways in patients with diabetes. lncRNA taurine upregulated gene 1 (TUG1) participates in angiogenesis in various cells. However, the mechanisms of TUG1 activity in EPCs have not been elucidated. METHODS We isolated and then characterized EPCs from the peripheral blood of mice using immunofluorescence and flow cytometry. Western blot detected the wnt/β-catenin pathway in high glucose-treated EPCs. Bioinformatics analysis predicted a putative binding site for TUG1 on miR-29c-3p. The interactions among TUG1, platelet-derived growth factor-BB (PDGF-BB), and miR-29c-3p were analyzed by luciferase assays. In vivo, diabetic mouse ischemic limb was treated with normal saline or TUG1 overexpression lentiviruses. RESULTS We found that EPC migration, invasion, and tube formation declined after treatment with high glucose, but improved with TUG1 overexpression. Mechanically, wnt/β-catenin pathway and autophagy were involved in the function of TUG1 overexpression in high glucose-treated EPCs. Moreover, TUG1 regulates the PDGF-BB/wnt pathway and function of high glucose-treated EPCs via miR-29c-3p. In vivo, injection of TUG1 lentivirus in a diabetic mouse ischemic limb model stimulated angiogenesis. CONCLUSIONS Our findings suggest that TUG1 restores high glucose-treated EPC function by regulating miR-29c-3p/PDGF-BB/Wnt signaling.
Collapse
Affiliation(s)
- Yang Li
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China
| | - Kangkang Zhi
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Shanghai, 200003, China
| | - Shilong Han
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China
| | - Xue Li
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China
| | - Maoquan Li
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China
| | - Weishuai Lian
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China.
| | - Haijun Zhang
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China.
| | - Xiaoping Zhang
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
43
|
Zhu J, Sun LL, Li WD, Li XQ. Clarification of the Role of miR-9 in the Angiogenesis, Migration, and Autophagy of Endothelial Progenitor Cells Through RNA Sequence Analysis. Cell Transplant 2020; 29:963689720963936. [PMID: 33028108 PMCID: PMC7784562 DOI: 10.1177/0963689720963936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We have previously reported that miR-9 promotes the homing, proliferation, and angiogenesis of endothelial progenitor cells (EPCs) by targeting transient receptor potential melastatin 7 via the AKT autophagy pathway. In this way, miR-9 promotes thrombolysis and recanalization following deep vein thrombosis (DVT). However, the influence of miR-9 on messenger RNA (mRNA) expression profiles of EPCs remains unclear. The current study comprises a comprehensive exploration of the mechanisms underlying the miR-9-regulated angiogenesis of EPCs and highlights potential treatment strategies for DVT. We performed RNA sequence analysis, which revealed that 4068 mRNAs were differentially expressed between EPCs overexpressing miR-9 and the negative control group, of which 1894 were upregulated and 2174 were downregulated. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses indicated that these mRNAs were mainly involved in regulating cell proliferation/migration processes/pathways and the autophagy pathway, both of which represent potential EPC-based treatment strategies for DVT. Reverse transcriptase quantitative polymerase chain reaction confirmed the changes in mRNA expression related to EPC angiogenesis, migration, and autophagy. We also demonstrate that miR-9 promotes EPC migration and angiogenesis by regulating FGF5 directly or indirectly. In summary, miR-9 enhances the expression of VEGFA, FGF5, FGF12, MMP2, MMP7, MMP10, MMP11, MMP24, and ATG7, which influences EPC migration, angiogenesis, and autophagy. We provide a comprehensive evaluation of the miR-9-regulated mRNA expression in EPCs and highlight potential targets for the development of new therapeutic interventions for DVT.
Collapse
Affiliation(s)
- Jian Zhu
- Department of Vascular Surgery, 105860The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Li-Li Sun
- Department of Vascular Surgery, 105860The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Vascular Surgery, Kunshan First People's Hospital, Kunshan, Jiangsu, China
| | - Wen-Dong Li
- Department of Vascular Surgery, Kunshan First People's Hospital, Kunshan, Jiangsu, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, 105860The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Vascular Surgery, Kunshan First People's Hospital, Kunshan, Jiangsu, China
| |
Collapse
|
44
|
Ghafouri-Fard S, Shoorei H, Mohaqiq M, Taheri M. Non-coding RNAs regulate angiogenic processes. Vascul Pharmacol 2020; 133-134:106778. [PMID: 32784009 DOI: 10.1016/j.vph.2020.106778] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis has critical roles in numerous physiologic processes during embryonic and adult life such as wound healing and tissue regeneration. However, aberrant angiogenic processes have also been involved in the pathogenesis of several disorders such as cancer and diabetes mellitus. Vascular endothelial growth factor (VEGF) is implicated in the regulation of this process in several physiologic and pathologic conditions. Notably, several non-coding RNAs (ncRNAs) have been shown to influence angiogenesis through modulation of expression of VEGF or other angiogenic factors. In the current review, we summarize the function and characteristics of microRNAs and long non-coding RNAs which regulate angiogenic processes. Understanding the role of these transcripts in the angiogenesis can facilitate design of therapeutic strategies to defeat the pathogenic events during this process especially in the human malignancies. Besides, angiogenesis-related mechanisms can improve tissue regeneration after conditions such as arteriosclerosis, myocardial infarction and limb ischemia. Thus, ncRNA-regulated angiogenesis can be involved in the pathogenesis of several disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Mohaqiq
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Liang H, Chen Y, Li H, Yu X, Xia C, Ming Z, Zhong C. miR-22-3p Suppresses Endothelial Progenitor Cell Proliferation and Migration via Inhibiting Onecut 1 (OC1)/Vascular Endothelial Growth Factor A (VEGFA) Signaling Pathway and Its Clinical Significance in Venous Thrombosis. Med Sci Monit 2020; 26:e925482. [PMID: 32876075 PMCID: PMC7486795 DOI: 10.12659/msm.925482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Proliferation and migration play crucial roles in various physiological processes, especially in injured endothelial repair. Endothelial progenitor cells (EPCs), as the precursors of endothelial cell, are involved in the regeneration of the endothelial lining of blood vessels. Furthermore, EPCs were found to be a potential choice for venous thrombosis (VT) treatment. Material/Methods EPCs were isolated from human peripheral blood of healthy adults and VT patients. Differently expressed micro(mi)RNAs were examined by quantitative real-time polymerase chain reaction, after which proliferative capacity and migration effect were tested by Cell-Counting Kit 8, scratch wound assay, and transwell assays. Bioinformatic analysis was applied to investigate the potential target messenger ribonucleic acid and a dual-luciferase reporting system was utilized to confirm the binding of miR-22-3p to its target gene. Western blot was carried out to detect candidate protein expression level. Finally, miR-22-3p expression was monitored in VT patients during follow-up to assess its correlation with prognosis of VT. Results Our data revealed that miR-22-3p was upregulated in EPCs derived from deep VT (DVT) individuals and suppression of miR-22-3p contributed to proliferation and migration of EPCs. In addition, miR-22-3p/onecut 1 (OC1)/vascular endothelial growth factor A (VEGFA) signaling pathway was involved in regulating EPC migration and proliferation. In addition, lower expression of miR-22-3p in DVT patients indicated decreased risk of VT recurrence. Conclusions Our results suggest that miR-22-3p regulates OC1/VEGFA signaling and is involved in regulating EPC proliferation and migration. The expression level of miR-22-3p could be monitored to predict DVT patients’ prognosis.
Collapse
Affiliation(s)
- Huoqi Liang
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Yibiao Chen
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Hefei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Xiaoqiang Yu
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Chunqiu Xia
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Zhibing Ming
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| | - Chongjun Zhong
- Department of Vascular Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China (mainland)
| |
Collapse
|
46
|
Zhao Z, Sun W, Guo Z, Zhang J, Yu H, Liu B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci 2020; 254:116900. [DOI: 10.1016/j.lfs.2019.116900] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022]
|
47
|
Metformin inhibits angiogenesis of endothelial progenitor cells via miR-221-mediated p27 expression and autophagy. Future Med Chem 2020; 11:2263-2272. [PMID: 31581911 DOI: 10.4155/fmc-2019-0017] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To explore the underlying mechanisms of metformin on the angiogenic capacity of endothelial progenitor cells (EPCs). Results: EPC growth and miR-221 expression decreased concentration-dependence with metformin, and a negative correlation was observed between miR-221 expression and metformin concentration (p < 0.001). miR-221 overexpression using a mimic decreased the metformin-mediated angiogenic effects in EPCs (p < 0.01). Metformin increased p27 and LC3II expression and AMP-activated protein kinase (AMPK) phosphorylation, and decreased p62 expression, while miR-221 overexpression reversed the effects of metformin. Additionally, AMPK inhibition by compound C reversed the increase in p27 and LC3II levels and AMPK phosphorylation or miR-221 siRNA treatment. Conclusion: Metformin inhibits the angiogenic capacity of EPCs. The underlying mechanism involves AMPK-mediated autophagy pathway activity and increases miR-221-mediated p27 expression.
Collapse
|
48
|
Cheng J, Hu W, Zheng F, Wu Y, Li M. hsa_circ_0058092 protects against hyperglycemia‑induced endothelial progenitor cell damage via miR‑217/FOXO3. Int J Mol Med 2020; 46:1146-1154. [PMID: 32705235 PMCID: PMC7387092 DOI: 10.3892/ijmm.2020.4664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) regulate the expression of genes that are critical for various biological and pathological processes. Previous studies have reported that the expression of hsa_circ_0058092 is decreased in patients with diabetes mellitus (DM); however, the specific role of this circRNA in DM is unknown. In the present study, endothelial progenitor cells (EPCs) were isolated and a decreased hsa_circ_0058092 expression was found under conditions of hyperglycemia (HG). The overexpression of hsa_circ_0058092 protected the EPCs against HG‑induced damage by preserving cell survival, proliferation, migration and angiogenic differentiation. The overexpression of hsa_circ_0058092 also decreased the HG‑induced increase in NADPH‑oxidase proteins and inflammatory cytokines. Further investigation revealed that the overexpression of hsa_circ_0058092 enhanced FOXO3 expression, which was mediated through the interaction with miR‑217. Furthermore, the upregulation of miR‑217 or the downregulation of FOXO3 abolished the protective effects of hsa_circ_0058092 against HG‑induced EPC damage. On the whole, these data suggest that hsa_circ_0058092 acts via the miR‑217/FOXO3 pathway to protect against EPCs HG‑induced damage, and to preserve the migration and angiogenesis of EPCs.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Guangzhou, Guangdong 510405, P.R. China
| | - Weiwei Hu
- Institute of Tropical Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Fenghui Zheng
- Department of Endocrinology and Metabolism, Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Yongfa Wu
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Guangzhou, Guangdong 510405, P.R. China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Guangzhou, Guangdong 510405, P.R. China
| |
Collapse
|
49
|
Zhang L, Jin C, Yang G, Wang B, Hua P, Zhang Y. LncRNA WTAPP1 promotes cancer cell invasion and migration in NSCLC by downregulating lncRNA HAND2-AS1. BMC Pulm Med 2020; 20:153. [PMID: 32473628 PMCID: PMC7260856 DOI: 10.1186/s12890-020-01180-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 05/11/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) Wilms Tumor 1 Associated Protein Pseudogene 1 (WTAPP1) has been reported to be a critical player in the angiogenesis and migration of endothelial progenitor cells, while its involvement in cancer biology remains unknown. This study was carried out to investigate the role of WTAPP1 in non-small cell lung cancer (NSCLC). METHODS The expression of WTAPP1 and lncRNA HAND2 Antisense RNA 1 (HAND2-AS1) in plasma and tissues from NSCLC patients was detected by qRT-PCR. A 5-year follow-up study was carried out to explore the prognostic value of WTAPP1 for NSCLC. Overexpression experiments were performed to analyze the interaction between WTAPP1 and HAND2-AS1. Cell invasion and migration were evaluated by Transwell assays. RESULTS The expression of WTAPP1 was upregulated in NSCLC. The survival analysis showed that low plasma levels of WTAPP1 were accompanied with high survival rate. HAND2-AS1 was downregulated in NSCLC and inversely correlated with WTAPP1 across tumor tissues. Overexpression of WTAPP1 resulted in downregulation of HAND2-AS1 in NSCLC cells, while overexpression of HAND2-AS1 did not affect the expression of WTAPP1. Overexpression of WTAPP1 led to promoted, while overexpression of HAND2-AS1 resulted in inhibited invasion and migration of NSCLC cells. In addition, overexpression of HAND2-AS1 partially attenuated the effects of overexpressing WTAPP1. In addition, WTAPP1 did not affect cancer cell proliferation. CONCLUSION WTAPP1 may promote cancer cell invasion and migration in NSCLC by downregulating lncRNA HAND2-AS1.
Collapse
Affiliation(s)
- Li Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun City, Jilin Province, 130041, People's Republic of China
| | - Chengyan Jin
- Department of Thoracic Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun City, Jilin Province, 130041, People's Republic of China
| | - Guiyun Yang
- Department of Operating Room, The Second Hospital of Jilin University, Changchun City, Jilin Province, 130041, People's Republic of China
| | - Bin Wang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun City, Jilin Province, 130041, People's Republic of China
| | - Peiyan Hua
- Department of Thoracic Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun City, Jilin Province, 130041, People's Republic of China
| | - Yan Zhang
- Department of Thoracic Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun City, Jilin Province, 130041, People's Republic of China.
| |
Collapse
|
50
|
Zhang Y, Sun J, Qi Y, Wang Y, Ding Y, Wang K, Zhou Q, Wang J, Ma F, Zhang J, Guo B. Long non-coding RNA TPT1-AS1 promotes angiogenesis and metastasis of colorectal cancer through TPT1-AS1/NF90/VEGFA signaling pathway. Aging (Albany NY) 2020; 12:6191-6205. [PMID: 32248186 PMCID: PMC7185097 DOI: 10.18632/aging.103016] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
LncRNAs have been proven closely correlated to tumor progression. A recent study identified LncRNA TPT1-AS1 (TPT1-AS1) as one of the liver-metastasis associated LncRNAs in colorectal cancer (CRC). In this study, we report that TPT1-AS1 is upregulated in CRC tissues, which is associated with poor prognosis. Functional assays unravel a pro-angiogenesis and metastasis role of TPT1-AS1. Mechanistically, Flexmap 3D assays reveal that TPT1-AS1 upregulates the VEGFA secretion in CRC cells. RNA immunoprecipitation and mRNA stability assays further show that TPT1-AS1 interacts with nuclear factor 90 (NF90) and subsequently promotes the association between NF90 and VEGFA mRNA, which leads to the upregulation of VEGFA mRNA stability. Therefore, we elucidate a new regulatory mechanism of TPT1-AS1 in CRC angiogenesis and targeting the TPT1-AS1/NF90/VEGFA axis may provide a useful strategy for diagnosis and treatment for colorectal cancer patients.
Collapse
Affiliation(s)
- Yiyun Zhang
- Department of Endoscopy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiangyun Sun
- Department of Acupuncture, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuan Qi
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yimin Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Ding
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kun Wang
- Department of Central Sterile Supply, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingxin Zhou
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jingxuan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fei Ma
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianguo Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Baoliang Guo
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|