1
|
Saadh MJ, Jasim NY, Ahmed MH, Ballal S, Kumar A, Atteri S, Vashishth R, Rizaev J, Alhili A, Jawad MJ, Yazdi F, Salajegheh A, Akhavan-Sigari R. Critical roles of miR-21 in promotions angiogenesis: friend or foe? Clin Exp Med 2025; 25:66. [PMID: 39998742 PMCID: PMC11861128 DOI: 10.1007/s10238-025-01600-7] [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: 11/09/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
MiRNAs are small RNA strands that are managed following transcription and are of substantial importance in blood vessel formation. It is essential to oversee the growth, differentiation, death, movement and construction of tubes by angiogenesis-affiliated cells. If miRNAs are not correctly regulated in regard to angiogenesis, it can deteriorate the health and lead to various illnesses, which include cancer, cardiovascular disorder, critical limb ischemia, Crohn's disease, ocular diseases, diabetic microvascular complications, and more. Consequently, it is vital to understand the crucial part that miRNAs play in the development of blood vessels, so we can develop reliable treatment plans for vascular diseases. This write-up will assess the critical role of miR-21/exosomal miR-21 in managing angiogenesis associated with bone growth, wound recovery, and other pathological conditions like tumor growth, ocular illnesses, diabetes, and other diseases connected to formation of blood vessels. Previous investigations have demonstrated that miR-21 is present at higher amounts in certain cancerous cells, and it influences a multitude of genes that moderate the increased creation of blood vessels. Furthermore, studies demonstrated that exosomal miR-21 has the capacity to interact with endothelial cells to foster tumor angiogenesis. For that reason, this review explains the critical importance of miR-21/exosomal miR-21 in managing both healthy and diseased states of angiogenesis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Nisreen Yasir Jasim
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Shikha Atteri
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | - Farzaneh Yazdi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Reza Akhavan-Sigari
- Dr. Schneiderhan GmbH and ISAR Klinikum, Munich, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw, Management University Warsaw, Warsaw, Poland
| |
Collapse
|
2
|
Jalouli M. Emerging Role of Hypoxia-Inducible Factors (HIFs) in Modulating Autophagy: Perspectives on Cancer Therapy. Int J Mol Sci 2025; 26:1752. [PMID: 40004215 PMCID: PMC11855875 DOI: 10.3390/ijms26041752] [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: 12/27/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are master regulators of cellular responses to low oxygen levels and modulate autophagy, a conserved process essential for maintaining homeostasis. Under hypoxic conditions, HIFs regulate the expression of autophagy-related genes and influence autophagic flux and cellular stress responses. Dysregulated hypoxia-induced autophagy promotes cancer cell survival, metabolism, and metastasis, thereby contributing to treatment resistance. Targeting HIF-mediated pathways or modulating autophagic processes offers the potential to improve traditional cancer therapies and overcome drug resistance. Pharmacological inhibitors of HIFs or autophagy, either alone or in combination with other treatments, may disrupt the pro-survival mechanisms within the hypoxic tumor microenvironment. Further research is needed to elucidate the intricate interplay between HIF signaling and the autophagy machinery in cancer cells. Understanding these processes could pave the way for novel therapeutic strategies to enhance treatment outcomes and combat drug resistance. This review highlights the complex relationship between HIFs and autophagy in cancer development and therapy, offering insights into how targeting these pathways may improve patient outcomes.
Collapse
Affiliation(s)
- Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| |
Collapse
|
3
|
Guo QY, Song JN, Chen YM, Yuan HN, Xue WS, Sun Y, Niu XL, Wang Y, Chen X. IL-6 regulates epithelial ovarian cancer EMT, invasion, and metastasis by modulating Let-7c and miR-200c through the STAT3/HIF-1α pathway. Med Oncol 2024; 41:155. [PMID: 38744773 DOI: 10.1007/s12032-024-02328-2] [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: 01/08/2024] [Accepted: 02/06/2024] [Indexed: 05/16/2024]
Abstract
Interleukin-6 (IL-6) and hypoxia-inducible factor-1α (HIF-1α) play important roles in epithelial-mesenchymal transformation (EMT) and tumor development. Previous studies have demonstrated that IL-6 promotes EMT, invasion, and metastasis in epithelial ovarian cancer (EOC) cells by activating the STAT3/HIF-1α pathway. MicroRNA (miRNA) is non-coding small RNAs that also play an important role in tumor development. Notably, Let-7 and miR-200 families are prominently altered in EOC. However, whether IL-6 regulates the expression of Let-7 and miR-200 families through the STAT3/HIF-1α signaling to induce EMT in EOC remains poorly understood. In this study, we conducted in vitro and in vivo investigations using two EOC cell lines, SKOV3, and OVCAR3 cells. Our findings demonstrate that IL-6 down-regulates the mRNA levels of Let-7c and miR-200c while up-regulating their target genes HMGA2 and ZEB1 through the STAT3/HIF-1α signaling in EOC cells and in vivo. Additionally, to explore the regulatory role of HIF-1α on miRNAs, both exogenous HIF blockers YC-1 and endogenous high expression or inhibition of HIF-1α can be utilized. Both approaches can confirm that the downstream molecule HIF-1α inhibits the expression and function of Let-7c and miR-200c. Further mechanistic research revealed that the overexpression of Let-7c or miR-200c can reverse the malignant evolution of EOC cells induced by IL-6, including EMT, invasion, and metastasis. Consequently, our results suggest that IL-6 regulates the expression of Let-7c and miR-200c through the STAT3/HIF-1α pathway, thereby promoting EMT, invasion, and metastasis in EOC cells.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/metabolism
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition/genetics
- Gene Expression Regulation, Neoplastic
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Interleukin-6/metabolism
- Interleukin-6/genetics
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/genetics
- Neoplasm Invasiveness/genetics
- Neoplasm Metastasis
- Neoplasms, Glandular and Epithelial/pathology
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/metabolism
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Signal Transduction
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
Collapse
Affiliation(s)
- Qiao Yun Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
- School of Pharmacy and Biological Technology, Tianjin Medical College, Tianjin, 300222, China
| | - Jiang Nan Song
- Department of Gynaecology and Obstetrics, Characteristic Medical Center of Chinese People's Armed Police Force, No.220, Chenglin Road, Dongli District, Tianjin, 300162, China
- Department of Gynecology and Obstetrics, Chinese People's Liberation Army General Hospital, Beijing, 100080, China
| | - Yu Meng Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hai Ning Yuan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Wen Shu Xue
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Yang Sun
- Department of Gynaecology and Obstetrics, Characteristic Medical Center of Chinese People's Armed Police Force, No.220, Chenglin Road, Dongli District, Tianjin, 300162, China
| | - Xiu Long Niu
- Institute of Prevention and Treatment of Dermatosis in Alpine Environment of Plateau, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, 300162, China
| | - Yue Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China.
| | - Xiao Chen
- Department of Gynaecology and Obstetrics, Characteristic Medical Center of Chinese People's Armed Police Force, No.220, Chenglin Road, Dongli District, Tianjin, 300162, China
| |
Collapse
|
4
|
Vatte S, Ugale R. HIF-1, an important regulator in potential new therapeutic approaches to ischemic stroke. Neurochem Int 2023; 170:105605. [PMID: 37657765 DOI: 10.1016/j.neuint.2023.105605] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide due to the narrow therapeutic window of the only approved therapies like intravenous thrombolysis and thrombectomy. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke by regulating multiple pathways including glucose metabolism, angiogenesis, neuronal survival, neuroinflammation and blood brain barrier regulation. Here, we give a brief overview of the HIF-1α-targeting strategies currently under investigation and summarise recent research on how HIF-1α is regulated in various brain cells, including neurons and microglia, at various stages in ischemic stroke. The roles of HIF-1 in stroke varies with ischemic time and degree of ischemia, are still up for debate. More focus has been placed on prospective HIF-1α targeting drugs, such as HIF-1α activator, HIF-1α stabilizers, and natural compounds. In this review, we have highlighted the regulation of HIF-1α in the novel therapeutic approaches for treatment of stroke.
Collapse
Affiliation(s)
- Sneha Vatte
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| |
Collapse
|
5
|
Jarosz-Popek J, Eyileten C, Gager GM, Nowak A, Szwed P, Wicik Z, Palatini J, von Lewinski D, Sourij H, Siller-Matula JM, Postula M. The interaction between non-coding RNAs and SGLT2: A review. Int J Cardiol 2023; 398:131419. [PMID: 39492411 DOI: 10.1016/j.ijcard.2023.131419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2, SLC5A2) is a promising target for a new class of drug primarily established as kidney-targeting as well as emerging class of glucose-lowering drugs in diabetes. Studies showed that SGLT2 inhibitors also have a systemic impact via indirectly targeting the heart and kidneys which exerts broad cardio- and nephroprotective effects. Additionally, as cancer cells tightly require glucose supply, studies also questioned how SGLT2 inhibitors impact molecular pathology and cellular metabolism in cancer hallmarks. However, the exact molecular mechanisms responsible for those benefits have not been fully discovered. MicroRNAs (miRNA) and circularRNAs (circRNAs) are endogenous, single-stranded, non-coding RNAs (ncRNAs) that can control protein-coding genes, affecting significant molecular and cellular processes regulating homeostasis. CircRNAs particularly regulate gene expression at the transcriptional and post-transcriptional level by sponging to miRNAs and by altering interactions between proteins.
Collapse
Affiliation(s)
- Joanna Jarosz-Popek
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland; Doctoral School, Medical University of Warsaw, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland; Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Gloria M Gager
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna 1090, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Anna Nowak
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland; Doctoral School, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Szwed
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland; Department of Neurochemistry, Institute of Psychiatry and Neurology, Sobieskiego 9 Street, Warsaw 02-957, Poland
| | - Jeff Palatini
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland; Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna 1090, Austria
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland.
| |
Collapse
|
6
|
Mafi A, Mannani R, Khalilollah S, Hedayati N, Salami R, Rezaee M, Dehmordi RM, Ghorbanhosseini SS, Alimohammadi M, Akhavan-Sigari R. The Significant Role of microRNAs in Gliomas Angiogenesis: A Particular Focus on Molecular Mechanisms and Opportunities for Clinical Application. Cell Mol Neurobiol 2023; 43:3277-3299. [PMID: 37414973 PMCID: PMC11409989 DOI: 10.1007/s10571-023-01385-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs with only 20-22 nucleic acids that inhibit gene transcription and translation by binding to mRNA. MiRNAs have a diverse set of target genes and can alter most physiological processes, including cell cycle checkpoints, cell survival, and cell death mechanisms, affecting the growth, development, and invasion of various cancers, including gliomas. So optimum management of miRNA expression is essential for preserving a normal biological environment. Due to their small size, stability, and capability of specifically targeting oncogenes, miRNAs have emerged as a promising marker and new biopharmaceutical targeted therapy for glioma patients. This review focuses on the most common miRNAs associated with gliomagenesis and development by controlling glioma-determining markers such as angiogenesis. We also summarized the recent research about miRNA effects on signaling pathways, their mechanistic role and cellular targets in the development of gliomas angiogenesis. Strategies for miRNA-based therapeutic targets, as well as limitations in clinical applications, are also discussed.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Mannani
- Department of Surgery, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Raziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Malihe Rezaee
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rohollah Mousavi Dehmordi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyedeh Sara Ghorbanhosseini
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tübingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
7
|
Sun J, Zhu S. Identifying the role of hypoxia-related lncRNAs in pancreatic cancer. Genomics 2023; 115:110665. [PMID: 37315872 DOI: 10.1016/j.ygeno.2023.110665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/30/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) play important roles in hypoxia-induced tumor processes. However, the prognostic value of hypoxia-related lncRNAs in pancreatic cancer is limited. METHODS Hypoxia-related lncRNAs were identified by coexpression analysis and the LncTarD database. LASSO analysis was performed to develop a prognostic model. The function of TSPOAP1-AS1 was studied in vitro and in vivo. RESULTS We recognized a set of fourteen hypoxia-related lncRNAs for the construction of a prognostic model. The prognostic model displayed excellent performance in predicting the prognosis of pancreatic cancer patients. The overexpression of TSPOAP1-AS1, a hypoxia-related lncRNA, attenuated the proliferation and invasion of pancreatic cancer cells. HIF-1α bound to the promoter of TSPOAP1-AS1 and impaired its transcription under hypoxia. CONCLUSION The hypoxia-related lncRNA assessment model might be a potential strategy for prognostic prediction in pancreatic cancer. The fourteen lncRNAs contained in the model could be helpful for uncovering the mechanisms of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Jing Sun
- The Stomatology Center of Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Academician workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shuai Zhu
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
8
|
Gao H, Nepovimova E, Heger Z, Valko M, Wu Q, Kuca K, Adam V. Role of hypoxia in cellular senescence. Pharmacol Res 2023; 194:106841. [PMID: 37385572 DOI: 10.1016/j.phrs.2023.106841] [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: 05/28/2023] [Revised: 06/25/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
Senescent cells persist and continuously secrete proinflammatory and tissue-remodeling molecules that poison surrounding cells, leading to various age-related diseases, including diabetes, atherosclerosis, and Alzheimer's disease. The underlying mechanism of cellular senescence has not yet been fully explored. Emerging evidence indicates that hypoxia is involved in the regulation of cellular senescence. Hypoxia-inducible factor (HIF)- 1α accumulates under hypoxic conditions and regulates cellular senescence by modulating the levels of the senescence markers p16, p53, lamin B1, and cyclin D1. Hypoxia is a critical condition for maintaining tumor immune evasion, which is promoted by driving the expression of genetic factors (such as p53 and CD47) while triggering immunosenescence. Under hypoxic conditions, autophagy is activated by targeting BCL-2/adenovirus E1B 19-kDa interacting protein 3, which subsequently induces p21WAF1/CIP1 as well as p16Ink4a and increases β-galactosidase (β-gal) activity, thereby inducing cellular senescence. Deletion of the p21 gene increases the activity of the hypoxia response regulator poly (ADP-ribose) polymerase-1 (PARP-1) and the level of nonhomologous end joining (NHEJ) proteins, repairs DNA double-strand breaks, and alleviates cellular senescence. Moreover, cellular senescence is associated with intestinal dysbiosis and an accumulation of D-galactose derived from the gut microbiota. Chronic hypoxia leads to a striking reduction in the amount of Lactobacillus and D-galactose-degrading enzymes in the gut, producing excess reactive oxygen species (ROS) and inducing senescence in bone marrow mesenchymal stem cells. Exosomal microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) play important roles in cellular senescence. miR-424-5p levels are decreased under hypoxia, whereas lncRNA-MALAT1 levels are increased, both of which induce cellular senescence. The present review focuses on recent advances in understanding the role of hypoxia in cellular senescence. The effects of HIFs, immune evasion, PARP-1, gut microbiota, and exosomal mRNA in hypoxia-mediated cell senescence are specifically discussed. This review increases our understanding of the mechanism of hypoxia-mediated cellular senescence and provides new clues for anti-aging processes and the treatment of aging-related diseases.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové 500 03, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno 613 00, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava 812 37, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou 434025, China; Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové 500 03, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové 500 03, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 500 05, Czech Republic; Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain.
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno 613 00, Czech Republic.
| |
Collapse
|
9
|
Gupta J, Tayyib NA, Jalil AT, Hlail SH, Zabibah RS, Vokhidov UN, Alsaikhan F, Ramaiah P, Chinnasamy L, Kadhim MM. Angiogenesis and prostate cancer: MicroRNAs comes into view. Pathol Res Pract 2023; 248:154591. [PMID: 37343381 DOI: 10.1016/j.prp.2023.154591] [Citation(s) in RCA: 4] [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: 05/15/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/23/2023]
Abstract
Angiogenesis, the formation of new blood vessels, is an important stage in the growth of cancer. Extracellular matrix, endothelial cells, and soluble substances must be carefully coordinated during the multistep procedure of angiogenesis. Inducers and inhibitors have been found to control pretty much every phase. In addition to benign prostatic hyperplasia, prostatic intraepithelial neoplasia, and angiogenesis have a critical role in the initiation and progression of prostate cancer. MicroRNA (miRNA) is endogenous, short, non-coding RNA molecules of almost 22 nucleotides play a role in regulating cellular processes and regulating several genes' expression. Through controlling endothelial migration, differentiation, death, and cell proliferation, miRNAs have a significant function in angiogenesis. A number of pathological and physiological processes, particularly prostate cancer's emergence, depend on the regulation of angiogenesis. Investigating the functions played with miRNAs in angiogenesis is crucial because it might result in the creation of novel prostate cancer therapies that entail regulating angiogenesis. The function of several miRNAs and its targeting genes engaged in cancer of the prostate angiogenesis will be reviewed in this review in light of the most recent developments. The potential clinical utility of miRNAs potentially a novel therapeutic targets will also be explored, as well as their capacity to control prostate cancer angiogenesis and the underlying mechanisms.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, U.P., India.
| | - Nahla A Tayyib
- Faculty of Nursing, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Hilla 51001, Babylon, Iraq.
| | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Ulug'bek N Vokhidov
- Department of ENT Diseases, Head of the Department of Quality Education, Tashkent State Dental Institute, Tashkent, Uzbekistan; Research scholar, Department of Scientific affairs, Samarkand State Medical Institute, Amir Temur Street 18, Samarkand, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | | | | | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad 10022 Iraq
| |
Collapse
|
10
|
Liu S, Kang L, Song Y, Miao M. Role of the HIF-1α/BNIP3 Signaling Pathway in Recurrent Hepatocellular Carcinoma and the Mechanism of Traditional Chinese Medicine. J Hepatocell Carcinoma 2023; 10:893-908. [PMID: 37313302 PMCID: PMC10259603 DOI: 10.2147/jhc.s409292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
Recurrence of hepatocellular carcinoma (HCC) negatively affects the quality of life of patients and leads to death. Studies have shown that recurrent hepatocellular carcinoma (RHCC) is closely related to tissue hypoxia and autophagy. It has been shown that hypoxia-inducible factor-1α (HIF-1α) and its downstream factor BCL-2 19 kDa-interacting protein 3 (BNIP3) promote cellular autophagy under hypoxic conditions, resulting in metastasis and RHCC. In this article, the molecular structures of HIF-1α and BNIP3 are described, and the significance of the HIF-1α/BNIP3 signaling pathway in RHCC is explained. Moreover, the role and mechanism of traditional Chinese medicine (TCM) in treating RHCC by modulating the HIF-1α/BNIP3 signaling pathway is discussed. Studies have shown that the HIF-1α/BNIP3 signaling pathway is a potential target of TCM in the treatment of RHCC. The mechanism of the HIF-1α/BNIP3 signaling pathway in RHCC and the progress achieved in TCM research on targeting and regulating this pathway are also reviewed in this article. The objective was to provide a theoretical basis for the prevention and treatment of RHCC, as well as further drug development.
Collapse
Affiliation(s)
- Sizhe Liu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, People’s Republic of China
| | - Le Kang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, People’s Republic of China
| | - Yagang Song
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, People’s Republic of China
| | - Mingsan Miao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, People’s Republic of China
| |
Collapse
|
11
|
Zeng CY, Wang XF, Hua FZ. HIF-1α in Osteoarthritis: From Pathogenesis to Therapeutic Implications. Front Pharmacol 2022; 13:927126. [PMID: 35865944 PMCID: PMC9294386 DOI: 10.3389/fphar.2022.927126] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis is a common age-related joint degenerative disease. Pain, swelling, brief morning stiffness, and functional limitations are its main characteristics. There are still no well-established strategies to cure osteoarthritis. Therefore, better clarification of mechanisms associated with the onset and progression of osteoarthritis is critical to provide a theoretical basis for the establishment of novel preventive and therapeutic strategies. Chondrocytes exist in a hypoxic environment, and HIF-1α plays a vital role in regulating hypoxic response. HIF-1α responds to cellular oxygenation decreases in tissue regulating survival and growth arrest of chondrocytes. The activation of HIF-1α could regulate autophagy and apoptosis of chondrocytes, decrease inflammatory cytokine synthesis, and regulate the chondrocyte extracellular matrix environment. Moreover, it could maintain the chondrogenic phenotype that regulates glycolysis and the mitochondrial function of osteoarthritis, resulting in a denser collagen matrix that delays cartilage degradation. Thus, HIF-1α is likely to be a crucial therapeutic target for osteoarthritis via regulating chondrocyte inflammation and metabolism. In this review, we summarize the mechanism of hypoxia in the pathogenic mechanisms of osteoarthritis, and focus on a series of therapeutic treatments targeting HIF-1α for osteoarthritis. Further clarification of the regulatory mechanisms of HIF-1α in osteoarthritis may provide more useful clues to developing novel osteoarthritis treatment strategies.
Collapse
Affiliation(s)
- Chu-Yang Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi-Feng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| | - Fu-Zhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| |
Collapse
|
12
|
Zhang M, Hu Y, Yang F, Zhang J, Zhang J, Yu W, Wang M, Lv X, Li J, Bai T, Chang F. Interaction between AhR and HIF-1 signaling pathways mediated by ARNT/HIF-1β. BMC Pharmacol Toxicol 2022; 23:26. [PMID: 35473600 PMCID: PMC9044668 DOI: 10.1186/s40360-022-00564-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 03/29/2022] [Indexed: 04/12/2024] Open
Abstract
Background The main causes of lung cancer are smoking, environmental pollution and genetic susceptibility. It is an indisputable fact that PAHs are related to lung cancer, and benzo(a) pyrene is a representative of PAHs. The purpose of the current investigation was to investigate the interaction between AhR and HIF-1 signaling pathways in A549 cells, which provide some experimental basis for scientists to find drugs that block AhR and HIF-1 signaling pathway to prevent and treat cancer. Methods This project adopts the CYP1A1 signaling pathways and the expression of CYP1B1 is expressed as a measure of AhR strength index. The expression of VEGF and CAIX volume as a measure of the strength of the signal path HIF-1 indicators. Through the construction of plasmid vector, fluorescence resonance energy transfer, real-time quantitative PCR, western blotting and immunoprecipitation, the interaction between AhR signaling pathway and HIF-1 signaling pathway was observed. Results BaP can enhance the binding ability of HIF-1α protein to HIF-1β/ARNT in a dose-dependent manner without CoCl2. However, the binding ability of AhR protein to HIF-1β/ARNT is inhibited by HIF-1α signaling pathway in a dose-dependent manner with CoCl2. Conclusion It is shown that activation of the AhR signaling pathway does not inhibit the HIF-1α signaling pathway, but activation of the HIF-1α signaling pathway inhibits the AhR signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00564-8.
Collapse
Affiliation(s)
- Mengdi Zhang
- Department of Pharmacy Experimental Teaching Center of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China.,Inner Mongolia Research Center for Drug Screening, Hohhot, China
| | - Yuxia Hu
- Inner Mongolia Research Center for Drug Screening, Hohhot, China.,The Center for New Drug Safety Evaluation and Research of Inner Mongolia Medical University, Hohhot, China
| | - Fan Yang
- School of Pharmaceutical Science, Shanxi Medical University, Hohhot, China
| | - Jingwen Zhang
- Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China
| | - Jianxin Zhang
- Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China
| | - Wanjia Yu
- Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China
| | - Minjie Wang
- Department of Pharmacology of Basic medical College, Inner Mongolia Medical university, Hohhot, China
| | - Xiaoli Lv
- Inner Mongolia Research Center for Drug Screening, Hohhot, China.,Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China
| | - Jun Li
- Inner Mongolia Research Center for Drug Screening, Hohhot, China.,The Center for New Drug Safety Evaluation and Research of Inner Mongolia Medical University, Hohhot, China
| | - Tuya Bai
- Inner Mongolia Research Center for Drug Screening, Hohhot, China. .,Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China.
| | - Fuhou Chang
- Inner Mongolia Research Center for Drug Screening, Hohhot, China. .,The Center for New Drug Safety Evaluation and Research of Inner Mongolia Medical University, Hohhot, China. .,Department of Pharmacology of Pharmaceutical College, Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
13
|
Jiang H, Zhao H, Zhang M, He Y, Li X, Xu Y, Liu X. Hypoxia Induced Changes of Exosome Cargo and Subsequent Biological Effects. Front Immunol 2022; 13:824188. [PMID: 35444652 PMCID: PMC9013908 DOI: 10.3389/fimmu.2022.824188] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/25/2022] [Indexed: 12/16/2022] Open
Abstract
Exosomes are small extracellular vesicles that are secreted by almost all types of cells and exist in almost all extracellular spaces. As an important mediator of intercellular communication, exosomes encapsulate the miRNA, lncRNA, cirRNA, mRNA, cytokine, enzyme, lipid, and other components from the cytoplasm into its closed single membrane structure and transfer them to recipient units in an autocrine, paracrine, or endocrine manner. Hypoxia is a state of low oxygen tension and is involved in many pathological processes. Hypoxia influences the size, quantity, and expression of exosome cargos. Exosomes derived from hypoxic tumor cells transfer genetics, proteins, and lipids to the recipient units to exert pleiotropic effects. Different donor cells produce different cargo contents, target different recipient units and lead to different biological effects. Hypoxic exosomes derived from tumor cells uptaken by normoxic tumor cells lead to promoted proliferation, migration, and invasion; uptaken by extracellular space or liver lead to promoted metastasis; uptaken by endothelial cells lead to promoted angiogenesis; uptaken by immune cells lead to promoted macrophage polarization and changed tumor immune microenvironment. In addition to various types of tumors, hypoxic exosomes also participate in the development of diseases in the cardiovascular system, neuron system, respiratory system, hematology system, endocrine system, urinary system, reproduction system, and skeletomuscular system. Understanding the special characteristics of hypoxic exosomes provide new insight into elaborating the pathogenesis of hypoxia related disease. This review summarizes hypoxia induced cargo changes and the biological effects of hypoxic exosomes in tumors and non-malignant diseases in different systems.
Collapse
Affiliation(s)
- Hongxia Jiang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Hanqiu Zhao
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Mengzhe Zhang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Yuanzhou He
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Xiaochen Li
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Yongjian Xu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases, National Ministry of Health of The People's Republic of China, Wuhan, China
| |
Collapse
|
14
|
Hypoxia-Induced circRNAs in Human Diseases: From Mechanisms to Potential Applications. Cells 2022; 11:cells11091381. [PMID: 35563687 PMCID: PMC9105251 DOI: 10.3390/cells11091381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/17/2022] Open
Abstract
Circular RNAs (circRNAs) are a special class of endogenous RNAs characterized by closed loop structures lacking 5′ to 3′ polarity and polyadenylated tails. They are widely present in various organisms and are more stable and conserved than linear RNAs. Accumulating evidence indicates that circRNAs play important roles in physiology-related processes. Under pathological conditions, hypoxia usually worsens disease progression by manipulating the microenvironment for inflammation and invasion through various dysregulated biological molecules. Among them, circRNAs, which are involved in many human diseases, including cancer, are associated with the overexpression of hypoxia-inducible factors. However, the precise mechanisms of hypoxic regulation by circRNAs remain largely unknown. This review summarizes emerging evidence regarding the interplay between circRNAs and hypoxia in the pathophysiological changes of diverse human diseases, including cancer. Next, the impact of hypoxia-induced circRNAs on cancer progression, therapeutic resistance, angiogenesis, and energy metabolism will be discussed. Last, but not least, the potential application of circRNAs in the early detection, prognosis, and treatment of various diseases will be highlighted.
Collapse
|
15
|
Circular RNA hsa_circ_0004543 Aggravates Cervical Cancer Development by Sponging MicroRNA hsa-miR-217 to Upregulate Hypoxia-Inducible Factor. JOURNAL OF ONCOLOGY 2022; 2022:4031403. [PMID: 35310917 PMCID: PMC8926462 DOI: 10.1155/2022/4031403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/29/2021] [Accepted: 01/10/2022] [Indexed: 11/21/2022]
Abstract
Cervical cancer (CC) is the 4th principal source of cancer death in females with 604,000 new patients and 342,000 deaths in 2020 worldwide. It has been extensively shown that circRNAs are involved in regulating CC development. Nevertheless, the function and mechanisms of hsa_circ_0004543 in regulating CC need to be clearly elucidated. Herein, hsa_circ_0004543 expressions were compared between 40 paired paracancerous and cancerous specimens from CC patients and between 6 CC cell lines and a normal human cervical epithelial cell line based on qRT-PCR. Potential complementary binding sites between hsa-miR-217 and hsa_circ_0004543 were predicted using the interactome, while binding sites for the hypoxia-inducible factor-1a (HIF-1a) were predicted by TargetScan. The function and mechanism of hsa_circ_0004543 in the development of CC were estimated by silencing hsa_circ_0004543 with/without hsa-miR-217 or HIF-1a overexpression. The association between gene expressions was evaluated with Pearson's correlation analysis. Molecular mechanisms were explored by ribonucleic acid (RNA) pulldown, dual-luciferase activity, and rescue experimental assays. Our results revealed that the hsa_circ_0004543 expression was considerably increased in CC tissues and cells. Its silencing repressed proliferation and metastasis, while it increased apoptosis of CC cells. The investigation of the mechanism showed that hsa-miR-217 silencing or HIF-1a overexpression rescued hsa_circ_0004543, and silencing inhibited malignant phenotypes of CC cells. hsa_circ_0004543 upregulated the HIF-1α expression by sponging hsa-miR-217 in CC development. Therefore, the hsa_circ_0004543 functioned as a competing endogenous RNA (ceRNA) of hsa-miR-217 to increase CC oncogenesis and metastasis by the upregulation of the HIF-1α expression. Consequently, targeting the hsa_circ_0004543/hsa-miR-217/HIF-1α axis might be a potential treatment approach for CC.
Collapse
|
16
|
He G, Peng X, Wei S, Yang S, Li X, Huang M, Tang S, Jin H, Liu J, Zhang S, Zheng H, Fan Q, Liu J, Yang L, Li H. Exosomes in the hypoxic TME: from release, uptake and biofunctions to clinical applications. Mol Cancer 2022; 21:19. [PMID: 35039054 PMCID: PMC8762953 DOI: 10.1186/s12943-021-01440-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a remarkable trait of the tumor microenvironment (TME). When facing selective pressure, tumor cells show various adaptive characteristics, such as changes in the expression of cancer hallmarks (increased proliferation, suppressed apoptosis, immune evasion, and so on) and more frequent cell communication. Because of the adaptation of cancer cells to hypoxia, exploring the association between cell communication mediators and hypoxia has become increasingly important. Exosomes are important information carriers in cell-to-cell communication. Abundant evidence has proven that hypoxia effects in the TME are mediated by exosomes, with the occasional formation of feedback loops. In this review, we equally focus on the biogenesis and heterogeneity of cancer-derived exosomes and their functions under hypoxia and describe the known and potential mechanism ascribed to exosomes and hypoxia. Notably, we call attention to the size change of hypoxic cancer cell-derived exosomes, a characteristic long neglected, and propose some possible effects of this size change. Finally, jointly considering recent developments in the understanding of exosomes and tumors, we describe noteworthy problems in this field that urgently need to be solved for better research and clinical application.
Collapse
Affiliation(s)
- Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shilei Tang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Sheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyu Zheng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
17
|
Hamidi AA, Zangoue M, Kashani D, Zangouei AS, Rahimi HR, Abbaszadegan MR, Moghbeli M. MicroRNA-217: a therapeutic and diagnostic tumor marker. Expert Rev Mol Diagn 2021; 22:61-76. [PMID: 34883033 DOI: 10.1080/14737159.2022.2017284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Cancer as one of the most common causes of death has always been one of the major health challenges globally. Since, the identification of tumors in the early tumor stages can significantly reduce mortality rates; it is required to introduce novel early detection tumor markers. MicroRNAs (miRNAs) have pivotal roles in regulation of cell proliferation, migration, apoptosis, and tumor progression. Moreover, due to the higher stability of miRNAs than mRNAs in body fluids, they can be considered as non-invasive diagnostic or prognostic markers in cancer patients. AREAS COVERED In the present review we have summarized the role of miR-217 during tumor progressions. The miR-217 functions were categorized based on its target molecular mechanisms and signaling pathways. EXPERT OPINION It was observed that miR-217 mainly exerts its function by regulation of the transcription factors during tumor progressions. The WNT, MAPK, and PI3K/AKT signaling pathways were also important molecular targets of miR-217 in different cancers. The present review clarifies the molecular biology of miR-217 and paves the way of introducing miR-217 as a non-invasive diagnostic marker and therapeutic target in cancer therapy.
Collapse
Affiliation(s)
- Amir Abbas Hamidi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Zangoue
- Department of Anesthesiology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Daniel Kashani
- Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Kaitsuka T, Matsushita M, Matsushita N. Regulation of Hypoxic Signaling and Oxidative Stress via the MicroRNA-SIRT2 Axis and Its Relationship with Aging-Related Diseases. Cells 2021; 10:cells10123316. [PMID: 34943825 PMCID: PMC8699081 DOI: 10.3390/cells10123316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/15/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
The sirtuin family of nicotinamide adenine dinucleotide-dependent deacetylase and ADP-ribosyl transferases plays key roles in aging, metabolism, stress response, and aging-related diseases. SIRT2 is a unique sirtuin that is expressed in the cytosol and is abundant in neuronal cells. Various microRNAs were recently reported to regulate SIRT2 expression via its 3'-untranslated region (UTR), and single nucleotide polymorphisms in the miRNA-binding sites of SIRT2 3'-UTR were identified in patients with neurodegenerative diseases. The present review highlights recent studies into SIRT2-mediated regulation of the stress response, posttranscriptional regulation of SIRT2 by microRNAs, and the implications of the SIRT2-miRNA axis in aging-related diseases.
Collapse
Affiliation(s)
- Taku Kaitsuka
- School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka 831-8501, Japan;
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan;
| | - Nobuko Matsushita
- Laboratory of Hygiene and Public Health, Department of Medical Technology, School of Life and Environmental Science, Azabu University, Sagamihara 252-5201, Japan
- Correspondence: ; Tel.: +81-42-769-1937
| |
Collapse
|
19
|
Mitroshina EV, Savyuk MO, Ponimaskin E, Vedunova MV. Hypoxia-Inducible Factor (HIF) in Ischemic Stroke and Neurodegenerative Disease. Front Cell Dev Biol 2021; 9:703084. [PMID: 34395432 PMCID: PMC8355741 DOI: 10.3389/fcell.2021.703084] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
Hypoxia is one of the most common pathological conditions, which can be induced by multiple events, including ischemic injury, trauma, inflammation, tumors, etc. The body's adaptation to hypoxia is a highly important phenomenon in both health and disease. Most cellular responses to hypoxia are associated with a family of transcription factors called hypoxia-inducible factors (HIFs), which induce the expression of a wide range of genes that help cells adapt to a hypoxic environment. Basic mechanisms of adaptation to hypoxia, and particularly HIF functions, have being extensively studied over recent decades, leading to the 2019 Nobel Prize in Physiology or Medicine. Based on their pivotal physiological importance, HIFs are attracting increasing attention as a new potential target for treating a large number of hypoxia-associated diseases. Most of the experimental work related to HIFs has focused on roles in the liver and kidney. However, increasing evidence clearly demonstrates that HIF-based responses represent an universal adaptation mechanism in all tissue types, including the central nervous system (CNS). In the CNS, HIFs are critically involved in the regulation of neurogenesis, nerve cell differentiation, and neuronal apoptosis. In this mini-review, we provide an overview of the complex role of HIF-1 in the adaptation of neurons and glia cells to hypoxia, with a focus on its potential involvement into various neuronal pathologies and on its possible role as a novel therapeutic target.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Savyuk
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Evgeni Ponimaskin
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
- Department of Cellular Neurophysiology, Hannover Medical School, Hanover, Germany
| | - Maria V. Vedunova
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
20
|
Tang Y, Zong S, Zeng H, Ruan X, Yao L, Han S, Hou F. MicroRNAs and angiogenesis: a new era for the management of colorectal cancer. Cancer Cell Int 2021; 21:221. [PMID: 33865381 PMCID: PMC8052662 DOI: 10.1186/s12935-021-01920-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/07/2021] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNA molecules containing only 20–22 nucleotides. MiRNAs play a role in gene silencing and translation suppression by targeting and binding to mRNA. Proper control of miRNA expression is very important for maintaining a normal physiological environment because miRNAs can affect most cellular pathways, including cell cycle checkpoint, cell proliferation, and apoptosis pathways, and have a wide range of target genes. With these properties, miRNAs can modulate multiple signalling pathways involved in cancer development, such as cell proliferation, apoptosis, and migration pathways. MiRNAs that activate or inhibit the molecular pathway related to tumour angiogenesis are common topics of research. Angiogenesis promotes tumorigenesis and metastasis by providing oxygen and diffusible nutrients and releasing proangiogenic factors and is one of the hallmarks of tumour progression. CRC is one of the most common tumours, and metastasis has always been a difficult issue in its treatment. Although comprehensive treatments, such as surgery, radiotherapy, chemotherapy, and targeted therapy, have prolonged the survival of CRC patients, the overall response is not optimistic. Therefore, there is an urgent need to find new therapeutic targets to improve CRC treatment. In a series of recent reports, miRNAs have been shown to bidirectionally regulate angiogenesis in colorectal cancer. Many miRNAs can directly act on VEGF or inhibit angiogenesis through other pathways (HIF-1a, PI3K/AKT, etc.), while some miRNAs, specifically many exosomal miRNAs, are capable of promoting CRC angiogenesis. Understanding the mechanism of action of miRNAs in angiogenesis is of great significance for finding new targets for the treatment of tumour angiogenesis. Deciphering the exact role of specific miRNAs in angiogenesis is a challenge due to the high complexity of their actions. Here, we describe the latest advances in the understanding of miRNAs and their corresponding targets that play a role in CRC angiogenesis and discuss possible miRNA-based therapeutic strategies.
Collapse
Affiliation(s)
- Yufei Tang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Shaoqi Zong
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.,Graduate School of Shanghai, University of Traditional Chinese Medicine, Shanghai, China
| | - Hailun Zeng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaofeng Ruan
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Liting Yao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Susu Han
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Fenggang Hou
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
21
|
Moghbeli M. Molecular interactions of miR-338 during tumor progression and metastasis. Cell Mol Biol Lett 2021; 26:13. [PMID: 33827418 PMCID: PMC8028791 DOI: 10.1186/s11658-021-00257-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer, as one of the main causes of human deaths, is currently a significant global health challenge. Since the majority of cancer-related deaths are associated with late diagnosis, it is necessary to develop minimally invasive early detection markers to manage and reduce mortality rates. MicroRNAs (miRNAs), as highly conserved non-coding RNAs, target the specific mRNAs which are involved in regulation of various fundamental cellular processes such as cell proliferation, death, and signaling pathways. MiRNAs can also be regulated by long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). They are highly stable in body fluids and have tumor-specific expression profiles, which suggest their suitability as efficient non-invasive diagnostic and prognostic tumor markers. Aberrant expression of miR-338 has been widely reported in different cancers. It regulates cell proliferation, migration, angiogenesis, and apoptosis in tumor cells. Main body In the present review, we have summarized all miR-338 interactions with other non-coding RNAs (ncRNAs) and associated signaling pathways to clarify the role of miR-338 during tumor progression. Conclusions It was concluded that miR-338 mainly functions as a tumor suppressor in different cancers. There were also significant associations between miR-338 and other ncRNAs in tumor cells. Moreover, miR-338 has a pivotal role during tumor progression using the regulation of WNT, MAPK, and PI3K/AKT signaling pathways. This review highlights miR-338 as a pivotal ncRNA in biology of tumor cells.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Patil N, Allgayer H, Leupold JH. MicroRNAs in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:1-31. [PMID: 33119862 DOI: 10.1007/978-3-030-50224-9_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is decisive for the eradication or survival of any tumor mass. Moreover, it plays a pivotal role for metastasis and for providing the metastatic niche. The TME offers special physiological conditions and is composed of, for example, surrounding blood vessels, the extracellular matrix (ECM), diverse signaling molecules, exosomes and several cell types including, but not being limited to, infiltrated immune cells, cancer-associated endothelial cells (CAEs), and cancer-associated fibroblasts (CAFs). These cells can additionally and significantly contribute to tumor and metastasis progression, especially also by acting via their own deregulated micro (mi) RNA expression or activity. Thus, miRNAs are essential players in the crosstalk between cancer cells and the TME. MiRNAs are small non-coding (nc) RNAs that typically inhibit translation and stability of messenger (m) RNAs, thus being able to regulate several cell functions including proliferation, migration, differentiation, survival, invasion, and several steps of the metastatic cascade. The dynamic interplay between miRNAs in different cell types or organelles such as exosomes, ECM macromolecules, and the TME plays critical roles in many aspects of cancer development. This chapter aims to give an overview on the multiple contributions of miRNAs as players within the TME, to summarize the role of miRNAs in the crosstalk between different cell populations found within the TME, and to illustrate how they act on tumorigenesis and the behavior of cells in the TME context. Lastly, the potential clinical utility of miRNAs for cancer therapy is discussed.
Collapse
Affiliation(s)
- Nitin Patil
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany
| | - Heike Allgayer
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany
| | - Jörg H Leupold
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany.
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karls University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
23
|
Chen L, Bao L, Niu Y, Wang JE, Kumar A, Xing C, Wang Y, Luo W. LncIHAT Is Induced by Hypoxia-Inducible Factor 1 and Promotes Breast Cancer Progression. Mol Cancer Res 2020; 19:678-687. [PMID: 33380467 DOI: 10.1158/1541-7786.mcr-20-0383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/03/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022]
Abstract
Hypoxia induces thousands of mRNAs and miRNAs to mediate tumor malignancy. However, hypoxia-induced long noncoding RNA (lncRNA) transcriptome and their role in triple-negative breast cancer (TNBC) have not been defined. Here we identified hypoxia-induced lncRNA transcriptome in two human TNBC cell lines by whole transcriptome sequencing. AC093818.1 was one of 26 validated lncRNAs and abundantly expressed in TNBC in vitro and in vivo. 5'- and 3'-rapid amplification of cDNA ends assays revealed that the isoform 2 was a dominant AC093818.1 transcript in TNBC cells and thus referred to as lncIHAT (lncRNA induced by hypoxia and abundant in TNBC). Hypoxia-inducible factor 1 (HIF1) but not HIF2 bound to the hypoxia response element at the promoter of lncIHAT to activate its transcription in hypoxic TNBC cells. LncIHAT promoted TNBC cell survival in vitro and tumor growth and lung metastasis in mice. Mechanistically, lncIHAT was required for the expression of its proximal neighboring oncogenic genes PDK1 and ITGA6 in TNBC cells and tumors. Reexpression of PDK1 and ITGA6 rescued survival and growth of lncIHAT knockdown TNBC cells in vitro. Collectively, these findings uncovered lncIHAT as a new hypoxia-induced oncogenic cis-acting lncRNA in TNBC. IMPLICATIONS: This study systematically identified hypoxia-induced lncRNA transcriptome in TNBC and sheds light on multiple layers of regulatory mechanisms of gene expression under hypoxia.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yanling Niu
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Jennifer E Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Ashwani Kumar
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas.,Department of Bioinformatics, UT Southwestern Medical Center, Dallas, Texas.,Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, Texas
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Neurology, UT Southwestern Medical Center, Dallas, Texas
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
24
|
Chen H, Zhang Y, Cao X, Mou P. MiR-27a Facilitates Breast Cancer Progression via GSK-3β. Technol Cancer Res Treat 2020; 19:1533033820965576. [PMID: 33025840 PMCID: PMC7545786 DOI: 10.1177/1533033820965576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Breast cancer remains one of the leading causes of cancer-associated death in women. MiR-27a is highly expressed in breast cancer tissue. However, the underlying mechanisms that promote breast cancer progression are unknown. In this study, we investigated the regulatory mechanisms of miR-27a and its target glycogen Synthase Kinase 3-β (GSK-3β) in breast cancer cells. We found that miR-27a was highly expressed in breast cancer tissues, which downregulated GSK-3β expression. We further identified GSK-3β as a direct target of miR-27a, and found that the miR-27a mediated suppression of GSK-3β activated Wnt/β-catenin-associated proliferative and invasive factor in breast cancer. The cell transfection assay demonstrated the overexpression of miR-27a also enhanced cell proliferation and invasion, and reduced cell apoptosis through GSK-3β. Finally, we demonstrated that the overexpression of miR-27a facilitated breast cancer progression through its ability to down-regulate the phosphorylation of GSK-3β both in vivo and vitro. These findings highlighted miR-27a as a novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Huijin Chen
- Department of Clinical Laboratory, ShengLi Oilfield Central Hospital, Dongying, Shandong, China
| | - Yuanyuan Zhang
- Department of Clinical Laboratory, ShengLi Oilfield Central Hospital, Dongying, Shandong, China
| | - Xin Cao
- Department of Orthopedic Trauma, ShengLi Oilfield Central Hospital, Dongying, Shandong, China
| | - Peipei Mou
- Department of Clinical Laboratory, ShengLi Oilfield Central Hospital, Dongying, Shandong, China
| |
Collapse
|
25
|
He J, Wang J, Li S, Li T, Chen K, Zhang S. Hypoxia-inhibited miR-338-3p suppresses breast cancer progression by directly targeting ZEB2. Cancer Sci 2020; 111:3550-3563. [PMID: 32726486 PMCID: PMC7540984 DOI: 10.1111/cas.14589] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Hypoxia plays an essential role in the development of various cancers. The biological function and underlying mechanism of microRNA-338-3p (miR-338-3p) under hypoxia remain unclarified in breast cancer (BC). Herein, we performed bioinformatics, gain and loss of function of miR-338-3p, a luciferase reporter assay, and chromatin immunoprecipitation (ChIP) in vitro and in a tumor xenograft model. We also explored the potential signaling pathways of miR-338-3p in BC. We detected the expression levels and prognostic significance of miR-338-3p in BC by qRT-PCR and in situ hybridization. MiR-338-3p was lowly expressed in BC tissues and cell lines, and BC patients with underexpression of miR-338-3p tend to have a dismal overall survival. Functional experiments showed that miR-338-3p overexpression inhibited BC cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) process, whereas miR-338-3p silencing abolished these biological behaviors. Zinc finger E-box-binding homeobox 2 (ZEB2) was validated as a direct target of miR-338-3p. ZEB2 overexpression promoted while ZEB2 knockdown abolished the promoted effects of miR-338-3p knockdown on cell biological behaviors through the NF-ĸB and PI3K/Akt signal pathways. HIF1A can transcriptionally downregulate miR-338-3p under hypoxia. In total, miR-338-3p counteracts hypoxia-induced BC cells growth, migration, invasion, and EMT via the ZEB2 and NF-ĸB/PI3K signal pathways, implicating miR-338-3p may be a promising target to treat patients with BC.
Collapse
Affiliation(s)
- Juanjuan He
- Department of Breast Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Jing Wang
- Department of Breast Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Songchao Li
- Department of Urology Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Teng Li
- Department of Urology Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Kunlun Chen
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| | - Shaojin Zhang
- Department of Urology Surgerythe First Affiliated HospitalZhengzhou UniversityZhengzhouChina
| |
Collapse
|
26
|
Huang H, Qing XY, Zhou Q, Li HD, Hu ZY. Silencing of microRNA-3175 represses cell proliferation and invasion in prostate cancer by targeting the potential tumor-suppressor SCN4B. Kaohsiung J Med Sci 2020; 37:20-26. [PMID: 32833340 DOI: 10.1002/kjm2.12292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/19/2020] [Accepted: 07/19/2020] [Indexed: 12/24/2022] Open
Abstract
MicroRNA-3175 (miR-3175) expression is upregulated in prostate cancer, but its roles and the underlying mechanisms in prostate cancer cell growth and invasion need to be elucidated. This study aimed to uncover the roles of miR-3175 in regulating cell growth and migration, as well as the expression of its predicted target gene cardiac sodium channel β4-subunit gene (SCN4B). Real-time quantitative PCR (RT-qPCR) and/or western blotting techniques were used to measure miR-3175 and SCN4B expression levels in prostate cancer cells. Inhibitor or mimics transfections were used to overexpress or silence miR-3175 in prostate cancer cells. MTT and Edu assays were applied to assess cell viability. Scratch assay and transwell chambers were used to examine cell migration and invasion abilities. The interaction between miR-3175 and SCN4B was determined by means of luciferase gene reporter, RT-qPCR, and western blotting assays. The results showed that miR-3175 expression was increased and SCN4B expression was decreased in prostate cancer cell lines as compared with normal human prostatic epithelial cells. Compared with the control group, knockdown of miR-3175 resulted in strong inhibitions of cell growth, migration, invasion, and N-cadherin expression, together with an increase in E-cadherin expression. In addition, knockdown of miR-3175 dramatically increased the luciferase activity of the luciferase vector of SCN4B, and increased SCN4B expression. Together, this study illustrated that downregulation of miR-3175 repressed the proliferation and invasion of prostate cancer cells, which might be induced by SCN4B downregulation.
Collapse
Affiliation(s)
- He Huang
- Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao-Yan Qing
- Department of Oncology, Chengdu Seventh People's Hospital, Chengdu, China
| | - Qiong Zhou
- Department of Medical, the Third Hospital of Nanchang, Nanchang, China
| | - Han-Dan Li
- Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhu-Yun Hu
- Department of Urology, the Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
27
|
Lu J, Chen H, He F, You Y, Feng Z, Chen W, Li X, Zhao L. Ginsenoside 20(S)-Rg3 upregulates HIF-1α-targeting miR-519a-5p to inhibit the Warburg effect in ovarian cancer cells. Clin Exp Pharmacol Physiol 2020; 47:1455-1463. [PMID: 32271958 DOI: 10.1111/1440-1681.13321] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 01/20/2023]
Abstract
The Warburg effect, one of the metabolic hallmarks of cancer, is responsible for rapid energy production through a high rate of aerobic glycolysis. Ginsenoside 20(S)-Rg3 antagonizes the Warburg effect in ovarian cancer cells by upregulating some microRNAs, including miR-519a-5p, that target key enzymes involved in aerobic glycolysis. How 20(S)-Rg3-upregulated miR-519a-5p influences the Warburg effect in ovarian cancer cells remains poorly defined, however. Here we report that while overexpression of miR-519a-5p in ovarian cancer cells inhibited the Warburg effect, inhibition of miR-519a-5p negated the suppressive action of 20(S)-Rg3 against the Warburg effect as evidenced by a decrease in glucose consumption, lactate production and HK2 expression. We identified HIF-1α as a direct target of miR-519a-5p via luciferase reporter assays and demonstrated the counteraction by overexpressed HIF-1α of 20(S)-Rg3-suppressed Warburg effect. Further, 20(S)-Rg3 decreased DNMT3A-mediated DNA methylation in the promoter region of its precursor gene, leading to an increase in the level of miR-519a-5p. In conclusion, 20(S)-Rg3 upregulates miR-519a-5p via reducing DNMT3A-mediated DNA methylation to inhibit HIF-1α-stimulated Warburg effect in ovarian cancer.
Collapse
Affiliation(s)
- Jiaojiao Lu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Fang He
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuanyi You
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhaozu Feng
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Chen
- Center for Laboratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Le Zhao
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
28
|
Liu X, Feng S, Zhang XD, Li J, Zhang K, Wu M, Thorne RF. Non-coding RNAs, metabolic stress and adaptive mechanisms in cancer. Cancer Lett 2020; 491:60-69. [PMID: 32726612 DOI: 10.1016/j.canlet.2020.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Abstract
Metabolic reprogramming in cancer describes the multifaceted alterations in metabolism that contribute to tumorigenesis. Major determinants of metabolic phenotypes are the changes in signalling pathways associated with oncogenic activation together with cues from the tumor microenvironment. Therein, depleted oxygen and nutrient levels elicit metabolic stress, requiring cancer cells to engage adaptive mechanisms. Non-coding RNAs (ncRNAs) act as regulatory elements within metabolic pathways and their widespread dysregulation in cancer contributes to altered metabolic phenotypes. Indeed, ncRNAs are the regulatory accomplices of many prominent effectors of metabolic reprogramming including c-MYC and HIFs that are activated by metabolic stress. By example, this review illustrates the range of ncRNAs mechanisms impacting these effectors throughout their DNA-RNA-protein lifecycle along with presenting the mechanistic roles of ncRNAs in adaptive responses to glucose, glutamine and lipid deprivation. We also discuss the facultative activation of metabolic enzymes by ncRNAs, a phenomenon which may reflect a broad but currently invisible level of metabolic regulation. Finally, the translational challenges associated with ncRNA discoveries are discussed, emphasizing the gaps in knowledge together with importance of understanding the molecular basis of ncRNA regulatory mechanisms.
Collapse
Affiliation(s)
- Xiaoying Liu
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Feng
- Key Laboratory of Regenerative Medicine, Ministry of Education, Department of Developmental & Regenerative Biology, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xu Dong Zhang
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Biomedical Sciences & Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Jinming Li
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China
| | - Kaiguang Zhang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China.
| | - Mian Wu
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230027, China; Key Laboratory of Stem Cell Differentiation & Modification, School of Clinical Medicine, Henan University, Zhengzhou, China.
| | - Rick F Thorne
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Molecular Pathology Centre, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450053, China; School of Environmental & Life Sciences, University of Newcastle, NSW, Australia.
| |
Collapse
|
29
|
Li Y, Wang Q, Ning N, Tang F, Wang Y. Bioinformatic analysis reveals MIR502 as a potential tumour suppressor in ovarian cancer. J Ovarian Res 2020; 13:77. [PMID: 32660514 PMCID: PMC7359466 DOI: 10.1186/s13048-020-00683-y] [Citation(s) in RCA: 4] [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: 05/08/2020] [Accepted: 07/07/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a major cause of death among women due to the lack of early screening methods and its complex pathological progression. Increasing evidence has indicated that microRNAs regulate gene expression in tumours by interacting with mRNAs. Although the research regarding OC and microRNAs is extensive, the vital role of MIR502 in OC remains unclear. METHODS We integrated two microRNA expression arrays from GEO to identify differentially expressed genes. The Kaplan-Meier method was used to screen for miRNAs that had an influence on survival outcome. Upstream regulators of MIR502 were predicted by JASPAR and verified by ChIP-seq data. The LinkedOmics database was used to study genes that were correlated with MIR502. Gene Set Enrichment Analysis (GSEA) was conducted for functional annotation with GO and KEGG pathway enrichment analyses by using the open access WebGestalt tool. We constructed a PPI network by using STRING to further explore the core proteins. RESULTS We found that the expression level of MIR502 was significantly downregulated in OC, which was related to poor overall survival. NRF1, as an upstream regulator of MIR502, was predicted by JASPAR and verified by ChIP-seq data. In addition, anti-apoptosis and pro-proliferation genes in the Hippo signalling pathway, including CCND1, MYC, FGF1 and GLI2, were negatively regulated by MIR502, as shown in the GO and KEGG pathway enrichment results. The PPI network further demonstrated that CCND1 and MYCN were at core positions in the development of ovarian cancer. CONCLUSIONS MIR502, which is regulated by NRF1, acts as a tumour suppressor gene to accelerate apoptosis and suppress proliferation by targeting the Hippo signalling pathway in ovarian cancer.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, China
| | - Qi Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, China
| | - Ning Ning
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, China
| | - Fanglan Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, China.
| |
Collapse
|
30
|
Zhou P, Xie W, Huang HL, Huang RQ, Tian C, Zhu HB, Dai YH, Li ZY. circRNA_100859 functions as an oncogene in colon cancer by sponging the miR-217-HIF-1α pathway. Aging (Albany NY) 2020; 12:13338-13353. [PMID: 32644049 PMCID: PMC7377858 DOI: 10.18632/aging.103438] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023]
Abstract
Circular RNAs (circRNAs) play an important role in cancer development and progression by regulating gene expression. The present study aimed to investigate the function of circRNA_100859 in colon cancer. circRNA expression profiles from a human circRNAs chip were analyzed. The effects of circRNA_100859 on cell proliferation and apoptosis were assessed in vitro and interactions between circRNA_100859 and its micro (mi)RNA and target genes were analyzed. The diagnostic and prognostic significance of circRNA_100859 was also investigated. It was identified that circRNA_100859 was overexpressed in colon cancer tissues and promoted cell proliferation and inhibited cell apoptosis. Additionally, bioinformatics and a dual-luciferase reporter assay confirmed that circRNA_100859 acted as a miR-217 sponge, and miR-217 directly targeted hypoxia-inducible factor (HIF)-1α. Rescue assays demonstrated that HIF-1α protein and mRNA expression levels and cell proliferation were regulated by the circRNA_100859/miR-217 axis (P<0.05). Furthermore, statistical analysis showed that the circRNA_100859-miR-217-HIF-1α axis was associated with Tumor-Node-Metastasis (TNM) stage, histological grade, and KRAS mutations, and also showed high diagnostic and prognostic value for patients with colon cancer (P<0.05). Therefore, it was concluded that circRNA_100859 functions as an oncogene in colon cancer by sponging the miR-217-HIF-1α pathway. In addition, the circRNA_100859-miR-217-HIF-1α axis may serve as a novel diagnostic and prognostic biomarker for patients with colon cancer.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.,Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xie
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Hua-Lin Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Rong-Qi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Chao Tian
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Hong-Bo Zhu
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying-Huan Dai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhi-Yuan Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China.,CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China.,GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
31
|
Guo F, Xue J. MicroRNA‑628‑5p inhibits cell proliferation and induces apoptosis in colorectal cancer through downregulating CCND1 expression levels. Mol Med Rep 2020; 21:1481-1490. [PMID: 32016467 PMCID: PMC7003041 DOI: 10.3892/mmr.2020.10945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/22/2019] [Indexed: 12/17/2022] Open
Abstract
MicroRNA (miR)-628-5p serves as an antitumor gene in a variety of cancers; however, the role of miR-628-5p in colorectal cancer remains largely unclear. The purpose of this study was to investigate the role and mechanism of miR-628-5p in colorectal cancer. Reverse transcription-quantitative PCR (RT-qPCR), colony formation assays and flow cytometric analysis were used to determine the expression levels of miR-628-5p in colorectal cancer tissues and cell lines, and the proliferative ability of colorectal cancer cells. TargetScan version 7.2 and dual-luciferase reporter assay were performed to predict and confirm miR-628-5p target genes. The expression levels of cyclin D1 (CCND1) and related genes were determined using RT-qPCR or/and western blotting analysis. miR-628-5p mimics and CCND1 plasmids were used to overexpress miR-628-5p and CCND1; it was demonstrated that the expression levels of miR-628-5p were significantly downregulated in colorectal cancer tissues and cell lines. miR-628-5p mimic-transfected cells inhibited the proliferation and induced apoptosis of HT-29 cells. CCND1, a downstream effector of miR-628-5p, promoted the proliferation and suppressed apoptosis of HT-29 cells, and the effects were reversed by miR-628-5p mimics. In conclusion, the present study suggested that colorectal cancer progression may be regulated through the miR-628-5p/CCND1 axis, and miR-628-5p could be used as a potential diagnostic and prognostic biomarker for colorectal cancer.
Collapse
Affiliation(s)
- Fei Guo
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075061, P.R. China
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075061, P.R. China
| |
Collapse
|
32
|
Qian W, Huang T, Feng W. Circular RNA HIPK3 Promotes EMT of Cervical Cancer Through Sponging miR-338-3p to Up-Regulate HIF-1α. Cancer Manag Res 2020; 12:177-187. [PMID: 32021434 PMCID: PMC6957911 DOI: 10.2147/cmar.s232235] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
Background Cervical cancer (CC) is the 4th most common malignancy and cancer-related fatality in women worldwide. Circular RNA is a newly recognized noncoding RNA form. More reports have confirmed their important regulatory functions in diverse physiological and cancer processes. However, the function and mechanisms of circ-HIPK3 in CC remain unknown. Methods circ-HIPK3 expression status was verified between 45 paired CC and normal adjacent tissues from 45 CC patients, also the 6 CC cell lines and a normal human cervical epithelial End1/E6E7 cell line by qRT-PCR. Effects of circ-HIPK3 silence on CC cell phenotypes were estimated. Circular RNA interactome was applied to forecast binding site between circ-HIPK3 and miRNAs. Pearson correlation analysis was used to confirm the relationship between genes. Point mutation, RNA pull-down, luciferase assay and rescue experiments were applied for molecular mechanism exploration. Results circ-HIPK3 expression was significantly elevated in CC cells and tissues. circ-HIPK3 silence repressed growth and metastasis, while induced apoptosis in CC cells. circ-HIPK3 sponged miRNA-338-3p (miR-338-3p); miR-338-3p to up-regulate hypoxia-inducible factor-1α (HIF-1α) and CC progress. MiR-338-3p silence or HIF-1α over-expression rescued circ-HIPK3 knockdown caused inhibition of CC malignant characteristics. Conclusion circ-HIPK3 acts as a competing endogenous RNA of miR-338-3p to promote cell growth and metastasis in CC, via regulating HIF-1α mediated EMT. Therefore, targeting circ-HIPK3/miR-338-3p/HIF-1α axis may be a novel therapeutic strategy for CC.
Collapse
Affiliation(s)
- Weiwei Qian
- Department of Gynecology, The First People's Hospital of Lianyungang, Lianyungang 222061, People's Republic of China.,Department of Gynecology, Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang 222061, People's Republic of China
| | - Tingting Huang
- Department of Gynecology, The First People's Hospital of Lianyungang, Lianyungang 222061, People's Republic of China.,Department of Gynecology, Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang 222061, People's Republic of China
| | - Wen Feng
- Department of Gynecology, The First People's Hospital of Lianyungang, Lianyungang 222061, People's Republic of China.,Department of Gynecology, Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang 222061, People's Republic of China
| |
Collapse
|
33
|
Jiang L, Yang Y, Lin Y, Chen Z, Xing C, Lu C, Yang H, Zhang S. An electrochemical sensor based on enzyme-free recycling amplification for sensitive and specific detection of miRNAs from cancer cells. Analyst 2020; 145:3353-3358. [DOI: 10.1039/d0an00275e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A catalyzed hairpin assembly and binding-induced formation of the DNA three-way junction for ultrasensitive electrochemical detection of diverse miRNAs is reported.
Collapse
Affiliation(s)
- Lili Jiang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Yuling Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Yuhong Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Ziyi Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Chao Xing
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Shusheng Zhang
- Collaborative Innovation Centre of Tumour Marker Detection Technology
- Equipment and Diagnosis-Therapy Integration in Universities of Shandong
- College of Chemistry and Chemical Engineering
- Linyi University
- Linyi 276005
| |
Collapse
|
34
|
Nuvoli B, Amadio B, Cortese G, Benedetti S, Antoniani B, Soriani A, Carosi M, Strigari L, Galati R. The effect of CELLFOOD TM on radiotherapy or combined chemoradiotherapy: preclinical evidence. Ther Adv Med Oncol 2019; 11:1758835919878347. [PMID: 31662796 PMCID: PMC6792276 DOI: 10.1177/1758835919878347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/04/2019] [Indexed: 11/16/2022] Open
Abstract
Background Based on previous observations that the nutraceutical CELLFOOD™ (CF), the 'physiological modulator' that aimed to make oxygen available 'on demand', inhibits the growth of cancer cells, this study was designed to investigate the role of CF in the regulation of hypoxia-inducible factor 1 alpha (HIF1α) and its correlated proteins, phosphoglycerate kinase 1 and vascular endothelial growth factor. Our idea was that CF, acting on HIF1α, in combination with current anticancer therapies could improve their effectiveness. Methods To evaluate the effect of CF in association with radiotherapy and chemotherapy, different human cancer cell lines and mice with mesothelioma were analysed by tumour growth, clonogenic assay, western blot and immunohistochemical analysis. Results CF in combination with radiation with or without cisplatin increases the death rate of cancer cells. In vivo, 70% of mice treated with CF before the mesothelioma graft did not show any tumour growth, indicating a possible preventive effect of CF. Moreover, in mouse mesothelioma xenografts, CF improves the effect of radiotherapy also in combination with chemotherapy treatment. Immunohistochemical analysis of tumour explants showed that HIF1α expression was reduced by the combination of CF and radiotherapy treatment and even more by the combination of CF and radiotherapy and chemotherapy treatment. Mechanistically, CF increases the fraction of oxygenated cells, making the radiotherapy more effective with a greater production of reactive oxygen species (ROS) that in turn, reduce the HIF1α expression. This effect is amplified by further increase in ROS from chemotherapy. Conclusions Collectively, results from preclinical trials suggest that CF could be a useful intervention to improve the efficacy of radiotherapy or combined treatment strategies and could be a promising treatment modality to counteract cancer.
Collapse
Affiliation(s)
- Barbara Nuvoli
- Preclinical Models and New Therapeutic Agent Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Bruno Amadio
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giancarlo Cortese
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Serena Benedetti
- Department of Biomolecular Sciences, University of Urbino 'Carlo Bo', Urbino, Italy
| | - Barbara Antoniani
- Anatomy Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Antonella Soriani
- Laboratory of Medical Physics and Expert Systems, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariantonia Carosi
- Anatomy Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lidia Strigari
- Laboratory of Medical Physics and Expert Systems, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Rossella Galati
- Preclinical Models and New Therapeutic Agent Unit, IRCCS Regina Elena National Cancer Institute, Via Chianesi, Rome 00144, Italy
| |
Collapse
|
35
|
Lv X, Li Y, Li Y, Li H, Zhou L, Wang B, Zhi Z, Tang W. FAL1: A critical oncogenic long non-coding RNA in human cancers. Life Sci 2019; 236:116918. [PMID: 31610208 DOI: 10.1016/j.lfs.2019.116918] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/29/2022]
Abstract
Long noncoding RNAs (lncRNAs) are characterized as a group of endogenous RNAs that are more than 200 nucleotides in length and have no protein-encoding function. More and more evidence indicates that lncRNAs play vital roles in various human diseases, especially in tumorigenesis. Focally amplified lncRNA on chromosome 1 (FAL1), a novel lncRNA with enhancer-like activity, has been identified as an oncogene in multiple cancers and high expression level of FAL1 is usually associated with poor prognosis. Dysregulation of FAL1 has been shown to promote the proliferation and metastasis of cancer cells. In the present review, we summarized and illustrated the functions and underlying molecular mechanisms of FAL1 in the occurrence and development of different cancers and other diseases. FAL1 has the potential to appear as a feasible diagnostic and prognostic tool and new therapeutic target for cancer patients though further investigation is needed so as to accelerate clinical application.
Collapse
Affiliation(s)
- Xiurui Lv
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuhan Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongxing Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Zhou
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Binyu Wang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengke Zhi
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
36
|
Mechanisms of hypoxia signalling: new implications for nephrology. Nat Rev Nephrol 2019; 15:641-659. [PMID: 31488900 DOI: 10.1038/s41581-019-0182-z] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
Studies of the regulation of erythropoietin (EPO) production by the liver and kidneys, one of the classical physiological responses to hypoxia, led to the discovery of human oxygen-sensing mechanisms, which are now being targeted therapeutically. The oxygen-sensitive signal is generated by 2-oxoglutarate-dependent dioxygenases that deploy molecular oxygen as a co-substrate to catalyse the post-translational hydroxylation of specific prolyl and asparaginyl residues in hypoxia-inducible factor (HIF), a key transcription factor that regulates transcriptional responses to hypoxia. Hydroxylation of HIF at different sites promotes both its degradation and inactivation. Under hypoxic conditions, these processes are suppressed, enabling HIF to escape destruction and form active transcriptional complexes at thousands of loci across the human genome. Accordingly, HIF prolyl hydroxylase inhibitors stabilize HIF and stimulate expression of HIF target genes, including the EPO gene. These molecules activate endogenous EPO gene expression in diseased kidneys and are being developed, or are already in clinical use, for the treatment of renal anaemia. In this Review, we summarize information on the molecular circuitry of hypoxia signalling pathways underlying these new treatments and highlight some of the outstanding questions relevant to their clinical use.
Collapse
|
37
|
Seo J, Han SY, Seong D, Han HJ, Song J. Multifaceted C-terminus of HSP70-interacting protein regulates tumorigenesis via protein quality control. Arch Pharm Res 2019; 42:63-75. [PMID: 30600426 DOI: 10.1007/s12272-018-1101-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 12/11/2018] [Indexed: 10/27/2022]
Abstract
C-terminus of heat shock protein 70 (HSP70)-interacting protein (CHIP) is an E3 ligase involved in a variety of protein homeostasis events implicated in diverse signaling pathways. Its involvement in varied and even opposite signaling circuits might be due to its hallmark signature of associating with molecular chaperones, including HSP90 and HSP70. Together, these proteins may be pivotal in implementing protein quality control. A curious and puzzling aspect of the function of CHIP is its capability to induce protein degradation via the proteasome- or lysosome-dependent pathways. In addition, these pathways are combined with ubiquitin-dependent or -independent pathways. This review focuses on the role of CHIP in the development or suppression of tumorigenesis. CHIP can act as a tumor suppressor by downregulating various oncogenes. CHIP also displays an oncogenic feature involving the inhibition of diverse tumor suppressors, including proteins related to intrinsic and extrinsic apoptotic pathways. The ability of CHIP to exhibit dual roles in determining the fate of cells has not been studied analytically. However, its association with various proteins involved in protein quality control might play a major role. In this review, the mechanistic roles of CHIP in tumor formation based on the regulation of diverse proteins are discussed.
Collapse
Affiliation(s)
- Jinho Seo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Su Yeon Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Daehyeon Seong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hyun-Ji Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|