1
|
Zhang Y, Wei Z, Xu H, Wang X, Gu T, Dong H, Chang H, Pang L. Identification of miR-451 target genes as prognostic markers in diffuse large B-cell lymphoma. Expert Rev Hematol 2024:1-12. [PMID: 39494860 DOI: 10.1080/17474086.2024.2422019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND B-cell lymphoma, a diverse malignancy, is intricately regulated by multiple factors. MicroRNAs (miRNAs) have been demonstrated to be important regulators of the initiation and progression of human B-cell lymphoma, but their functions need to be further explored. RESEARCH DESIGN AND METHODS Two B-cell lymphoma cell lines, Romas and HBL-1, were engineered to overexpress miR-451, with cell proliferation assessed via cell counting assays. Flow cytometry was used to study the effects of miR-451 on cell cycle and apoptosis. Bioinformatics analyses were performed using GEO datasets (GSE181063, GSE32918, GSE56315) to identify DEGs, and potential miR-451 target genes were predicted using tools like ENCORI, TargetScan, and R packages. A risk model for DLBCL prognosis was developed using Cox and LASSO regression. qRT-PCR validated the expression of these target genes. RESULTS This study revealed that miR-451 inhibited cell proliferation, arrested the cell cycle, and induced apoptosis in human DLBCL cell lines. Bioinformatics analysis identified 9 target genes (MMP9, AQP9, RIN2, EOMES, LCP2, SELPLG, MAL, SOCS5, S1PR3) significantly associated with DLBCL prognosis, suggesting a potential mechanism by which miR-451 suppresses DLBCL development. CONCLUSIONS Our study indicates that a specific set of miR-451 target genes may significantly influence DLBCL patient outcomes.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
- Department of Clinical Pharmacy, The People's Hospital of Jiaozuo, Jiaozuo, Henan, China
| | - Zichen Wei
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Han Xu
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Xin Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Ting Gu
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Hongliang Dong
- Department of Clinical Pharmacy, The People's Hospital of Jiaozuo, Jiaozuo, Henan, China
| | - Hongzhuan Chang
- Department of Clinical Pharmacy, The People's Hospital of Jiaozuo, Jiaozuo, Henan, China
| | - Lei Pang
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Eftekhari M, Safari A, Najafi M. MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome. Cancer Cell Int 2024; 24:233. [PMID: 38965615 PMCID: PMC11229485 DOI: 10.1186/s12935-024-03420-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/22/2024] [Indexed: 07/06/2024] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate genes and are involved in various biological processes, including cancer development. Researchers have been exploring the potential of miRNAs as therapeutic agents in cancer treatment. Specifically, targeting the mammalian target of the rapamycin (mTOR) pathway with miRNAs has shown promise in improving the effectiveness of radiotherapy (RT), a common cancer treatment. This review provides an overview of the current understanding of miRNAs targeting mTOR as therapeutic agents to enhance RT outcomes in cancer patients. It emphasizes the importance of understanding the specific miRNAs that target mTOR and their impact on radiosensitivity for personalized cancer treatment approaches. The review also discusses the role of mTOR in cell homeostasis, cell proliferation, and immune response, as well as its association with oncogenesis. It highlights the different ways in which miRNAs can potentially affect the mTOR pathway and their implications in immune-related diseases. Preclinical findings suggest that combining mTOR modulators with RT can inhibit tumor growth through anti-angiogenic and anti-vascular effects, but further research and clinical trials are needed to validate the efficacy and safety of using miRNAs targeting mTOR as therapeutic agents in combination with RT. Overall, this review provides a comprehensive understanding of the potential of miRNAs targeting mTOR to enhance RT efficacy in cancer treatment and emphasizes the need for further research to translate these findings into improved clinical outcomes.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Seyed Mohammad Amini
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Eftekhari
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Safari
- Department of Radiology, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, 71439-14693, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
4
|
Hu P, Cao Q, Feng H, Liu Y, Chen Y, Xu J, Feng W, Sun H, Ding H, Wang C, Gao J, Xiao M. MicroRNA-451a is a candidate biomarker and therapeutic target for major depressive disorder. Gen Psychiatr 2024; 37:e101291. [PMID: 38304710 PMCID: PMC10831421 DOI: 10.1136/gpsych-2023-101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024] Open
Abstract
Background Increasing evidence supports the role of microRNAs (miRNAs) in major depressive disorder (MDD), but the pathophysiological mechanism remains elusive. Aims To explore the mechanism of microRNA-451a (miR-451a) in the pathology and behaviours of depression. Methods Abnormal miRNAs such as miR-451a reported previously in the serum of patients with MDD were screened and then confirmed in a mouse model of depression induced by chronic restraint stress (CRS). Eight-week-old male C57BL/6 mice had miR-451a overexpression in the medial prefrontal cortex (mPFC) via adeno-associated virus serotype 9 vectors encoding a pri-mmu-miR-451a-GFP fusion protein followed by behavioural and pathological analyses. Finally, molecular biological experiments were conducted to investigate the potential mechanism of miR-451a against depression. Results The serum levels of miRNA-451a were significantly lower in patients with MDD, with a negative correlation with the Hamilton Depression Scale scores. Additionally, a negative association between serum miR-451a and behavioural despair or anhedonia was observed in CRS mice. Notably, miR-451a expression was significantly downregulated in the mPFC of CRS-susceptible mice. Overexpressing miR-451a in the mPFC reversed the loss of dendritic spines and the depression-like phenotype of CRS mice. Mechanistically, miR-451a could inhibit CRS-induced corticotropin-releasing factor receptor 1 expression via targeting transcription factor 2, subsequently protecting dendritic spine plasticity. Conclusions Together, these results highlighted miR-451a as a candidate biomarker and therapeutic target for MDD.
Collapse
Affiliation(s)
- Panpan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiuchen Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hu Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Liu
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingfan Xu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huaiqing Sun
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huachen Ding
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Takahashi K, Tsuji M, Nakagawasai O, Katsuyama S, Hong L, Miyagawa K, Kurokawa K, Mochida-Saito A, Takeda H, Tadano T. Donepezil prevents olfactory dysfunction and α-synuclein aggregation in the olfactory bulb by enhancing autophagy in zinc sulfate-treated mice. Behav Brain Res 2023; 438:114175. [PMID: 36309244 DOI: 10.1016/j.bbr.2022.114175] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/18/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease is associated with marked olfactory dysfunction observed in the early stages. Clinical studies reported that acetylcholinesterase inhibitor donepezil (DNP) attenuated this deficit; however, the underlying mechanism remains unclear. Herein, we aimed to examine the effects and underlying mechanisms of DNP on olfactory deficits in zinc sulfate (ZnSO4) nasal-treated mice, which were used as a model of reversible olfactory impairment. We evaluated olfactory function using the buried food finding test and neurogenesis in the subventricular zone (SVZ) using immunohistochemistry. Finally, we measured the expression of doublecortin (DCX), neuronal nuclear antigen (NeuN), olfactory marker protein, tyrosine hydroxylase (TH), tryptophan hydroxylase 2, glutamic acid decarboxylase 67, p-α-synuclein (Ser129), α-synuclein, p-AMPK, p-p70S6 kinase (p70S6K) (Thr389), LC3 Ⅱ/Ⅰ, and p-p62 in the olfactory bulb (OB) by western blotting. On day 7 after treatment, ZnSO4-treated mice exhibited prolonged time to find the buried food, cell proliferation enhancement in the SVZ, increased NeuN, p-α-synuclein (Ser129), and α-synuclein levels, and decreased DCX and TH levels in the OB; except for TH, these changes normalized on day 14 after treatment. Repeated administration of DNP prevented the ZnSO4-induced changes on day 7 after treatment. Moreover, DNP increased p-AMPK and LC3 Ⅱ/Ⅰ, and decreased p-p70S6K and p-p62 (Ser351) levels in the OB, suggesting that DNP enhances autophagy in the OB. These findings indicate that DNP may help prevent olfactory dysfunction by autophagy that reduces α-synuclein aggregation via the AMPK/mTOC1 pathway.
Collapse
Affiliation(s)
- Kohei Takahashi
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Minoru Tsuji
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan.
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| | - Soh Katsuyama
- Division of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, 10281 Komuro, Kitaadachigun Inamachi, Saitama 362-0806, Japan
| | - Lihua Hong
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuya Miyagawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Kazuhiro Kurokawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Atsumi Mochida-Saito
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, Tochigi 324-8501, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa, Fukuoka 831-8501, Japan
| | - Takeshi Tadano
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan; Department of Environment and Preventive Medicine, Graduate School of Medicine Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
6
|
Epi-miRNAs: Regulators of the Histone Modification Machinery in Human Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4889807. [PMID: 35087589 PMCID: PMC8789461 DOI: 10.1155/2022/4889807] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of death and disability worldwide. Epigenetic deregulation is one of the most critical mechanisms in carcinogenesis and can be classified into effects on DNA methylation and histone modification. MicroRNAs are small noncoding RNAs involved in fine-tuning their target genes after transcription. Various microRNAs control the expression of histone modifiers and are involved in a variety of cancers. Therefore, overexpression or downregulation of microRNAs can alter cell fate and cause malignancies. In this review, we discuss the role of microRNAs in regulating the histone modification machinery in various cancers, with a focus on the histone-modifying enzymes such as acetylases, deacetylases, methyltransferases, demethylases, kinases, phosphatases, desumoylases, ubiquitinases, and deubiquitinases. Understanding of microRNA-related aberrations underlying histone modifiers in pathogenesis of different cancers can help identify novel therapeutic targets or early detection approaches that allow better management of patients or monitoring of treatment response.
Collapse
|
7
|
Yang W, Wei X, Jiao Y, Bai Y, Sam WN, Yan Q, Sun X, Li G, Ma J, Wei W, Tian D, Zheng F. STAT3/HIF-1α/fascin-1 axis promotes RA FLSs migration and invasion ability under hypoxia. Mol Immunol 2021; 142:83-94. [PMID: 34971867 DOI: 10.1016/j.molimm.2021.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) synovium was identified as "tumor-like" tissues because of the hypoxic microenvironment, significant cell proliferation, and invasion phenotypes. It was reported that hypoxia promoted tumor aggressiveness via up-regulated expression of fascin-1 in cancer. However, the role of fascin-1 in RA synovial hyperplasia and joint injury progression remains unknown. In the current study, we first identified that both fascin-1 and HIF-1α were highly expressed in the RA synovium, in which they were widely colocalized, compared to osteoarthritis(OA). As well, levels of fascin-1 in RA fibroblast-like synoviocytes(FLSs) were found significantly higher than those in OA FLSs. Further, it was demonstrated that the mRNA and protein levels of fascin-1 in RA FLSs were up-regulated in hypoxia (3 % O2) and experimental hypoxia induced by cobalt chloride. Mechanistically, the HIF-1α-mediated hypoxia environment activated the gene expression of the fascin-1 protein, which in turn promoted the migration and invasion of RA FLSs. Accordingly, the restoration of FLSs migration and invasion was observed following siRNA-mediated silencing of fascin-1 and HIF-1α expression. Notably, under the experimental hypoxia, we found that the expression levels of fascin-1, HIF-1α, and p-STAT3 were increased in a time-dependent manner, and fascin-1and HIF-1α expressions were dependent on p-STAT3. Our results indicated that hypoxia-induced fascin-1 up-regulation promoted RA FLSs migration and invasion through the STAT3/HIF-1α/fascin-1 axis, which might represent a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Wang Yang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xinyue Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yachong Jiao
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Hebei, China
| | - Yingyu Bai
- Laboratory for Mechanisms and Therapies of Heart Diseases, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wilfried Noel Sam
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Qiushuang Yan
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China.
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
8
|
Yi YW, You KS, Park JS, Lee SG, Seong YS. Ribosomal Protein S6: A Potential Therapeutic Target against Cancer? Int J Mol Sci 2021; 23:ijms23010048. [PMID: 35008473 PMCID: PMC8744729 DOI: 10.3390/ijms23010048] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Ribosomal protein S6 (RPS6) is a component of the 40S small ribosomal subunit and participates in the control of mRNA translation. Additionally, phospho (p)-RPS6 has been recognized as a surrogate marker for the activated PI3K/AKT/mTORC1 pathway, which occurs in many cancer types. However, downstream mechanisms regulated by RPS6 or p-RPS remains elusive, and the therapeutic implication of RPS6 is underappreciated despite an approximately half a century history of research on this protein. In addition, substantial evidence from RPS6 knockdown experiments suggests the potential role of RPS6 in maintaining cancer cell proliferation. This motivates us to investigate the current knowledge of RPS6 functions in cancer. In this review article, we reviewed the current information about the transcriptional regulation, upstream regulators, and extra-ribosomal roles of RPS6, with a focus on its involvement in cancer. We also discussed the therapeutic potential of RPS6 in cancer.
Collapse
Affiliation(s)
- Yong Weon Yi
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
| | - Seok-Geun Lee
- Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.-G.L.); (Y.-S.S.); Tel.: +82-2-961-2355 (S.-G.L.); +82-41-550-3875 (Y.-S.S.); Fax: +82-2-961-9623 (S.-G.L.)
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
- Correspondence: (S.-G.L.); (Y.-S.S.); Tel.: +82-2-961-2355 (S.-G.L.); +82-41-550-3875 (Y.-S.S.); Fax: +82-2-961-9623 (S.-G.L.)
| |
Collapse
|
9
|
Gupta I, Vranic S, Al-Thawadi H, Al Moustafa AE. Fascin in Gynecological Cancers: An Update of the Literature. Cancers (Basel) 2021; 13:cancers13225760. [PMID: 34830909 PMCID: PMC8616296 DOI: 10.3390/cancers13225760] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Fascin, an actin-binding protein, is upregulated in different types of human cancers. It is reportedly responsible for increasing the invasive and metastatic ability of cancer cells by reducing cell–cell adhesions. This review provides a brief overview of fascin and its interactions with other genes and oncoviruses to induce the onset and progression of cancer. Abstract Fascin is an actin-binding protein that is encoded by the FSCN1 gene (located on chromosome 7). It triggers membrane projections and stimulates cell motility in cancer cells. Fascin overexpression has been described in different types of human cancers in which its expression correlated with tumor growth, migration, invasion, and metastasis. Moreover, overexpression of fascin was found in oncovirus-infected cells, such as human papillomaviruses (HPVs) and Epstein-Barr virus (EBV), disrupting the cell–cell adhesion and enhancing cancer progression. Based on these findings, several studies reported fascin as a potential biomarker and a therapeutic target in various cancers. This review provides a brief overview of the FSCN1 role in various cancers with emphasis on gynecological malignancies. We also discuss fascin interactions with other genes and oncoviruses through which it might induce cancer development and progression.
Collapse
Affiliation(s)
- Ishita Gupta
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Semir Vranic
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Hamda Al-Thawadi
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
| | - Ala-Eddin Al Moustafa
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (I.G.); (S.V.); (H.A.-T.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar
- Biomedical Research Centre, QU Health, Qatar University, Doha 2713, Qatar
- Correspondence: ; Tel.: +974-4403-7817
| |
Collapse
|
10
|
Estradiol ameliorates metformin-inhibited Sertoli cell proliferation via AMPK/TSC2/mTOR signaling pathway. Theriogenology 2021; 175:7-22. [PMID: 34481229 DOI: 10.1016/j.theriogenology.2021.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/31/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022]
Abstract
Metformin is a commonly used for treating type 2 diabetes and it acts on a variety of organs including the male reproductive system. 17β-estradiol plays an important role in Sertoli cell (SC) proliferation which determines the germ cell development and spermatogenesis. The aim of this study is to investigate the effect of metformin on immature chicken SC proliferation and the potential mechanisms by which 17β-estradiol regulate this process. Results showed that metformin significantly inhibited SC proliferation, whereas 17β-estradiol weakened the inhibitory effects of metformin on SC viability, cell growth, and cell cycle progression. SC proliferation-inhibiting effect of metformin exposure was regulated by decreasing adenosine triphosphate level and respiratory enzyme activity in the mitochondria; this process was possibly mediated by the adenosine monophosphate-activated protein kinase (AMPK)/tuberous sclerosis complex 2 (TSC2)/mammalian target of rapamycin (mTOR) signaling pathway, which was regulated by the down-expressed miR-1764 and by the decreased antioxidant enzyme activity and excessive reactive oxygen species generation. In addition, SCs transfected with the miR-1764 agomir led to an improvement of proliferation capacity through down-regulating AMPKα2 level, which further decreased TSC2 expression and induced mTOR activation. However, the anti-proliferative effect of miR-1764 antagomir can be alleviated by 17β-estradiol treatment via the up-expression of miR-1764 in transfected SCs. Our findings suggest appropriate dose of exogenous 17β-estradiol treatment can ameliorate the inhibitory effect of metformin on SC proliferation via the regulation of AMPK/TSC2/mTOR signaling pathway, this might reduce the risk of poor male fertility caused by the abuse of anti-diabetic agents.
Collapse
|
11
|
Ristic B, Kopel J, Sherazi SAA, Gupta S, Sachdeva S, Bansal P, Ali A, Perisetti A, Goyal H. Emerging Role of Fascin-1 in the Pathogenesis, Diagnosis, and Treatment of the Gastrointestinal Cancers. Cancers (Basel) 2021; 13:cancers13112536. [PMID: 34064154 PMCID: PMC8196771 DOI: 10.3390/cancers13112536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Gastrointestinal (GI) cancers, including esophageal, gastric, colorectal, liver, and pancreatic cancers, remain as one of the leading causes of death worldwide, with a large proportion accounting for fatalities related to metastatic disease. The active involvement of fascin-1 in forming membrane protrusions crucial for cellular movement has been identified as an important molecular mechanism behind the phenotypic switch from the localized to the metastatic tumor. Thus, fascin-1 expression status in the malignant tissue has been utilized as an important component in determining the patient’s clinicopathological outcomes. In this review, we provide an up-to-date literature review of the role of fascin-1 in the initiation and metastatic progression of GI tract cancers, its involvement in patients’ clinical outcomes, and its potential as a therapeutic target. Abstract Gastrointestinal (GI) cancers, including esophageal, gastric, colorectal, liver, and pancreatic cancers, remain as one of the leading causes of death worldwide, with a large proportion accounting for fatalities related to metastatic disease. Invasion of primary cancer occurs by the actin cytoskeleton remodeling, including the formation of the filopodia, stereocilia, and other finger-like membrane protrusions. The crucial step of actin remodeling in the malignant cells is mediated by the fascin protein family, with fascin-1 being the most active. Fascin-1 is an actin-binding protein that cross-links filamentous actin into tightly packed parallel bundles, giving rise to finger-like cell protrusions, thus equipping the cell with the machinery necessary for adhesion, motility, and invasion. Thus, fascin-1 has been noted to be a key component for determining patient diagnosis and treatment plan. Indeed, the overexpression of fascin-1 in GI tract cancers has been associated with a poor clinical prognosis and metastatic progression. Moreover, fascin-1 has received attention as a potential therapeutic target for metastatic GI tract cancers. In this review, we provide an up-to-date literature review of the role of fascin-1 in the initiation of GI tract cancers, metastatic progression, and patients’ clinical outcomes.
Collapse
Affiliation(s)
- Bojana Ristic
- Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Jonathan Kopel
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Syed A. A. Sherazi
- Department of Medicine, John H Stroger Jr Hospital of Cook County, Chicago, IL 60612, USA;
| | - Shweta Gupta
- Division of Hematology-Oncology, John H Stroger Jr Hospital of Cook County, Chicago, IL 60612, USA;
| | - Sonali Sachdeva
- Department of Cardiology, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Pardeep Bansal
- Department of Gastroenterology, Mercy Health-St. Vincent Medical Center, Toledo, OH 43608, USA;
| | - Aman Ali
- Department of Medicine, The Commonwealth Medical College, Scranton, PA 18510, USA;
| | - Abhilash Perisetti
- Department of Gastroenterology and Hepatology, The University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Hemant Goyal
- The Wright Center for Graduate Medical Education, Scranton, PA 18510, USA
- Correspondence:
| |
Collapse
|
12
|
Liu H, Zhang Y, Li L, Cao J, Guo Y, Wu Y, Gao W. Fascin actin-bundling protein 1 in human cancer: promising biomarker or therapeutic target? Mol Ther Oncolytics 2021; 20:240-264. [PMID: 33614909 PMCID: PMC7873579 DOI: 10.1016/j.omto.2020.12.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fascin actin-bundling protein 1 (FSCN1) is a highly conserved actin-bundling protein that cross links F-actin microfilaments into tight, parallel bundles. Elevated FSCN1 levels have been reported in many types of human cancers and have been correlated with aggressive clinical progression, poor prognosis, and survival outcomes. The overexpression of FSCN1 in cancer cells has been associated with tumor growth, migration, invasion, and metastasis. Currently, FSCN1 is recognized as a candidate biomarker for multiple cancer types and as a potential therapeutic target. The aim of this study was to provide a brief overview of the FSCN1 gene and protein structure and elucidate on its actin-bundling activity and physiological functions. The main focus was on the role of FSCN1 and its upregulatory mechanisms and significance in cancer cells. Up-to-date studies on FSCN1 as a novel biomarker and therapeutic target for human cancers are reviewed. It is shown that FSCN1 is an unusual biomarker and a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Hongliang Liu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yu Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Li Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Wei Gao
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| |
Collapse
|
13
|
Yan W, Wang Y, Chen Y, Guo Y, Li Q, Wei X. Exosomal miR-130b-3p Promotes Progression and Tubular Formation Through Targeting PTEN in Oral Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:616306. [PMID: 33829013 PMCID: PMC8019696 DOI: 10.3389/fcell.2021.616306] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC), accounting for two-thirds of head and neck cancer, is characterized by poor prognosis and a high rate of mortality. Exosomes have emerged as potential molecule-shuttle in intercellular communication, thereby regulating the physiological processes of recipient cells. To date, the effect of exosomal microRNAs (miRNAs) on the progression of OSCC has not been fully investigated. In this study, we found that the protein, but not mRNA expression of Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was decreased in OSCC. The results revealed that miR-130b-3p was an important negative regulator for PTEN expression. Additionally, overexpression and knockdown of miR-130b-3p enhanced and inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs), respectively. Also, miR-130b-3p was transferred by exosomes to HUVECs and then promoted angiogenesis and inhibit the expression of PTEN. Furthermore, exosomal miR-130b-3p derived from OSCC cells promoted tumor growth and blood vessel formation in the xenograft mice model. Taken together, we demonstrated that exosome-mediated miR-130b-3p promoted progression and tubular formation in OSCC in vitro and in vivo. These results would provide new insight into exploring biomarkers and effective therapeutic strategies for OSCC.
Collapse
Affiliation(s)
- Wei Yan
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yuping Wang
- Department of Stomatology of Shennongju Hospital, Huanghua, China
| | - Yong Chen
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yanjun Guo
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Li
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaotong Wei
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
14
|
Niknafs B, Hesam Shariati MB, Shokrzadeh N. miR223-3p, HAND2, and LIF expression regulated by calcitonin in the ERK1/2-mTOR pathway during the implantation window in the endometrium of mice. Am J Reprod Immunol 2021; 85:e13333. [PMID: 32869441 DOI: 10.1111/aji.13333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/11/2020] [Accepted: 08/22/2020] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Approximately one-third of infertility cases are related to the female partner, and implantation failure is the primary reason for female infertility. The current research was established to assess the impact of calcitonin on endometrial receptivity. METHODS OF STUDY 64 female BALB/c mice were assigned to 2 groups as follows: mice with regular ovarian cycle and mice with stimulated ovarian cycle. The two groups were further divided into four subgroups as follows: control (Ctrl), calcitonin (CT), pp242, and CT + pp242 groups. Calcitonin and pp242 were injected on days 3, 4, and 5 of pregnancy. On day 5 of gestation, all of the animals were sacrificed, and their uterine was removed for the morphological analysis, as well as the expression assessment genes and proteins. RESULTS The results demonstrated that ovarian stimulation increased the rate of phosphorylation of ERK1/2 and mTOR proteins, and resulted in the upregulation of miR-223-3p. The administration of calcitonin also elevated the expression levels of LIF and HAND2 gene in both regular ovarian and ovarian-stimulated cycles. In ovarian-stimulated groups, the administration of calcitonin led to a decrease in the expression of miR-223-3p. Calcitonin administration also markedly increased the phosphorylation of 4EBP1 and ERK1/2 in the regular ovarian cycle. CONCLUSION It seems that calcitonin is capable of enhancing the endometrial receptivity of the uterine, thereby the overexpression of HAND2 and LIF and downregulation of miR-223-3p through the ERK1/2-mTOR signaling pathway.
Collapse
Affiliation(s)
- Behrooz Niknafs
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Naser Shokrzadeh
- Department of Anatomical Sciences, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
15
|
Ait-Aissa K, Nguyen QM, Gabani M, Kassan A, Kumar S, Choi SK, Gonzalez AA, Khataei T, Sahyoun AM, Chen C, Kassan M. MicroRNAs and obesity-induced endothelial dysfunction: key paradigms in molecular therapy. Cardiovasc Diabetol 2020; 19:136. [PMID: 32907629 PMCID: PMC7488343 DOI: 10.1186/s12933-020-01107-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/28/2020] [Indexed: 01/17/2023] Open
Abstract
The endothelium plays a pivotal role in maintaining vascular health. Obesity is a global epidemic that has seen dramatic increases in both adult and pediatric populations. Obesity perturbs the integrity of normal endothelium, leading to endothelial dysfunction which predisposes the patient to cardiovascular diseases. MicroRNAs (miRNAs) are short, single-stranded, non-coding RNA molecules that play important roles in a variety of cellular processes such as differentiation, proliferation, apoptosis, and stress response; their alteration contributes to the development of many pathologies including obesity. Mediators of obesity-induced endothelial dysfunction include altered endothelial nitric oxide synthase (eNOS), Sirtuin 1 (SIRT1), oxidative stress, autophagy machinery and endoplasmic reticulum (ER) stress. All of these factors have been shown to be either directly or indirectly caused by gene regulatory mechanisms of miRNAs. In this review, we aim to provide a comprehensive description of the therapeutic potential of miRNAs to treat obesity-induced endothelial dysfunction. This may lead to the identification of new targets for interventions that may prevent or delay the development of obesity-related cardiovascular disease.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Quynh My Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, USA
| | - Mohanad Gabani
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Adam Kassan
- Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, Los Angeles, USA
| | - Santosh Kumar
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Soo-Kyoung Choi
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia, Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Tahsin Khataei
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Amal M Sahyoun
- Department of Food Science and Agriculture Chemistry, McGill University, Montreal, QC, Canada
| | - Cheng Chen
- Department of emergency and Critical Care, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Modar Kassan
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
16
|
miR-107 inhibition upregulates CAB39 and activates AMPK-Nrf2 signaling to protect osteoblasts from dexamethasone-induced oxidative injury and cytotoxicity. Aging (Albany NY) 2020; 12:11754-11767. [PMID: 32527986 PMCID: PMC7343481 DOI: 10.18632/aging.103341] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
To human osteoblasts dexamethasone (DEX) treatment induces significant oxidative injury and cytotoxicity. Inhibition of CAB39 (calcium binding protein 39)-targeting microRNA can induce CAB39 upregulation, activating AMP-activated protein kinase (AMPK) signaling and offering osteoblast cytoprotection. Here we identified a novel CAB39-targeting miRNA: the microRNA-107 (miR-107). RNA-Pull down assay results demonstrated that the biotinylated-miR-107 directly binds to CAB39 mRNA in OB-6 human osteoblastic cells. Forced overexpression of miR-107, by infection of pre-miR-107 lentivirus or transfection of wild-type miR-107 mimic, largely inhibited CAB39 expression in OB-6 cells and primary human osteoblasts. Contrarily, miR-107 inhibition, by antagomiR-107, increased its expression, resulting in AMPK cascade activation. AntagomiR-107 largely attenuated DEX-induced cell death and apoptosis in OB-6 cells and human osteoblasts. Importantly, osteoblast cytoprotection by antagomiR-107 was abolished with AMPK in-activation by AMPKα1 dominant negative mutation, silencing or knockout. Further studies demonstrated that antagomiR-107 activated AMPK downstream Nrf2 cascade to inhibit DEX-induced oxidative injury. Conversely, Nrf2 knockout almost abolished antagomiR-107-induced osteoblast cytoprotection against DEX. Collectively, miR-107 inhibition induced CAB39 upregulation and activated AMPK-Nrf2 signaling to protect osteoblasts from DEX-induced oxidative injury and cytotoxicity.
Collapse
|
17
|
Jin C, Zhao W, Zhang Z, Liu W. Silencing circular RNA circZNF609 restrains growth, migration and invasion by up-regulating microRNA-186-5p in prostate cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3350-3358. [PMID: 31387394 DOI: 10.1080/21691401.2019.1648281] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: Prostate cancer (PC) fearfully impacts men's health. We explored the efficacy and mechanism of circular RNA circZNF609 (circZNF609) on colony formation, viability, apoptosis, migration and invasion and in PC cells. Methods: Colony formation, CCK-8, flow cytometry, migration and invasion assay were respectively used to detect the functions of circZNF609 and microRNA (miR)-186-5p on cell colony ability, viability, apoptosis, migration and invasion. circZNF609 and miR-186-5p expression were changed by cell transfection and tested by RT-qPCR. Moreover, Cleaved-Caspase-3, Cleaved-Caspase-9, matrix metalloproteinase-9 (MMP-9), Vimentin and relate-proteins of cell pathways were examined through Western blot. Results: circZNF609 was highly expressed at PC tissues. circZNF609 declined cell colony ability, viability, migration and invasion and caused apoptosis. Furthermore, circZNF609 negatively regulated miR-186-5p, miR-186-5p inhibitor could reverse impacts of circZNF609. Finally, circZNF609 restrained the YAP1 and AMPK pathways by up-regulating miR-186-5p. Conclusion: Silencing circZNF609 restrained growth, migration and invasion of PC cells by up-regulating miR-186-5p via YAP1 and AMPK pathways. Highlights circZNF609 is highly expressed in PC tissues; circZNF609 restrains cell growth, migration and invasion; circZNF609 exerts its function by up-regulating miR-186-5p; circZNF609 exerts its function by YAP1 and AMPK signaling pathways.
Collapse
Affiliation(s)
- Chengjun Jin
- a Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Weiming Zhao
- a Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Zijian Zhang
- a Department of Urology, The First Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Wanpeng Liu
- b Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University , Harbin , China
| |
Collapse
|
18
|
Xu F, Liu Y, Zhu X, Li S, Shi X, Li Z, Ai M, Sun J, Hou B, Cai W, Sun H, Ni L, Zhou Y, Qiu L. Protective Effects and Mechanisms of Vaccarin on Vascular Endothelial Dysfunction in Diabetic Angiopathy. Int J Mol Sci 2019; 20:ijms20184587. [PMID: 31533227 PMCID: PMC6769517 DOI: 10.3390/ijms20184587] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are a major leading cause of mortality in patients suffering from type 2 diabetes mellitus (T2DM). Vascular endothelial dysfunction is a core pathophysiological event in the early stage of T2DM and eventually leads to cardiovascular disease. Vaccarin (VAC), an active flavonoid glycoside extracted from vaccariae semen, exhibits extensive biological activities including vascular endothelial cell protection effects. However, little is known about whether VAC is involved in endothelial dysfunction regulation under high glucose (HG) or hyperglycemia conditions. Here, in an in vivo study, we found that VAC attenuated increased blood glucose, increased glucose and insulin tolerance, relieved the disorder of lipid metabolism and oxidative stress, and improved endothelium-dependent vasorelaxation in STZ/HFD-induced T2DM mice. Furthermore, in cultured human microvascular endothelial cell-1 (HMEC-1) cells, we showed that pretreatment with VAC dose-dependently increased nitric oxide (NO) generation and the phosphorylation of eNOS under HG conditions. Mechanistically, VAC-treated HMEC-1 cells exhibited higher AMPK phosphorylation, which was attenuated by HG stimulation. Moreover, HG-triggered miRNA-34a upregulation was inhibited by VAC pretreatment, which is in accordance with pretreatment with AMPK inhibitor compound C (CC). In addition, both reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC) and VAC abolished HG-evoked dephosphorylation of AMPK and eNOS, increased miRNA-34a expression, and decreased NO production. These results suggest that VAC impedes HG-induced endothelial dysfunction via inhibition of the ROS/AMPK/miRNA-34a/eNOS signaling cascade.
Collapse
Affiliation(s)
- Fei Xu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Yixiao Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Xuexue Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Shuangshuang Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Xuelin Shi
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Zhongjie Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Min Ai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Jiangnan Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Lulu Ni
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Yuetao Zhou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214100, China.
| |
Collapse
|
19
|
Sun Y, Lei B, Huang Q. SOX18 Affects Cell Viability, Migration, Invasiveness, and Apoptosis in Hepatocellular Carcinoma (HCC) Cells by Participating in Epithelial-to-Mesenchymal Transition (EMT) Progression and Adenosine Monophosphate Activated Protein Kinase (AMPK)/Mammalian Target of Rapamycin (mTOR). Med Sci Monit 2019; 25:6244-6254. [PMID: 31427562 PMCID: PMC6713035 DOI: 10.12659/msm.915729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common malignancies around the world. It has been verified that the expression of SOX18 is correlated to poor clinical prognosis in patients with ovarian cancer, non-small cell lung cancer, or breast invasive ductal carcinoma. However, the expression pattern and biological function of SOX18 in HCC tissues remains unclear. In this study, we set out to investigate the associated biological function and potential molecular mechanism of the SOX18 gene in HCC cells. Material/Methods The mRNA and protein expression levels of experimental related genes were detected by real-time polymerase chain reaction and western blotting assay, respectively. The MTT method was used to assess cell viability, and cell apoptosis analysis was performed by means of FACScan flow cytometry. Wound-healing assay and Transwell analysis were performed to evaluate the ability of cell migration and invasiveness, respectively. Results SOX18 was highly expressed in various HCC cell lines. In addition, SOX18 promoted cell viability, migration, and invasion and simultaneously induce cell apoptosis. SOX18 promoted epithelial-to-mesenchymal transition (EMT) progression, and SOX18 downregulation activated the autophagy signaling pathway AMPK/mTOR in HCC cells. Conclusions SOX18 downregulation in HCC cells suppressed cell viability and metastasis, induced cell apoptosis and hindered the occurrence and progression of tumor cells by participating in the EMT process and regulating the autophagy signaling pathway AMPK/mTOR.
Collapse
Affiliation(s)
- Yanni Sun
- Department of Hepatology, Yantai City Hospital for Infectious Diseases, Yantai, Shandong, China (mainland)
| | - Bo Lei
- Department of Hepatology, Yantai City Hospital for Infectious Diseases, Yantai, Shandong, China (mainland)
| | - Qingxian Huang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China (mainland)
| |
Collapse
|
20
|
Javid H, Soltani A, Mohammadi F, Hashemy SI. Emerging roles of microRNAs in regulating the mTOR signaling pathway during tumorigenesis. J Cell Biochem 2019; 120:10874-10883. [PMID: 30719752 DOI: 10.1002/jcb.28401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/13/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a large Ser/Thr protein kinase that belongs to the phosphoinositide 3-kinase (PI3K) family and mediates various physiological and pathological processes, especially cell proliferation, protein synthesis, autophagy, and cancer development. The mTOR expression is transient and tightly regulated in normal cells, but it is overactivated in cancer cells. Recently, several studies have indicated that microRNAs (miRNAs) play a critical role in the regulation of mTOR and mTOR-associated processes, some acting as inhibitors and the others as activators. Although it is still in infancy, the strategy of combining both miRNAs and mTOR inhibitors might provide an approach to selectively sensitizing tumor cells to chemotherapy-induced DNA damage and subsequently attenuating the tumor cell growth and apoptosis.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Soltani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Mohammadi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Zhou Y, Hu Y, Sun Q, Xie N. Non-coding RNA in Fragile X Syndrome and Converging Mechanisms Shared by Related Disorders. Front Genet 2019; 10:139. [PMID: 30881383 PMCID: PMC6405884 DOI: 10.3389/fgene.2019.00139] [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: 10/14/2018] [Accepted: 02/11/2019] [Indexed: 12/12/2022] Open
Abstract
Fragile X syndrome (FXS) is one of the most common forms of hereditary intellectual disability. It is also a well-known monogenic cause of autism spectrum disorders (ASD). Repetitive trinucleotide expansion of CGG repeats in the 5'-UTR of FMR1 is the pathological mutation. Full mutation CGG repeats epigenetically silence FMR1 and thus lead to the absence of its product, fragile mental retardation protein (FMRP), which is an indispensable translational regulator at synapsis. Loss of FMRP causes abnormal neural morphology, dysregulated protein translation, and distorted synaptic plasticity, giving rise to FXS phenotypes. Non-coding RNAs, including siRNA, miRNA, and lncRNA, are transcribed from DNA but not meant for protein translation. They are not junk sequence but play indispensable roles in diverse cellular processes. FXS is the first neurological disorder being linked to miRNA pathway dysfunction. Since then, insightful knowledge has been gained in this field. In this review, we mainly focus on how non-coding RNAs, especially the siRNAs, miRNAs, and lncRNAs, are involved in FXS pathogenesis. We would also like to discuss several potential mechanisms mediated by non-coding RNAs that may be shared by FXS and other related disorders.
Collapse
Affiliation(s)
- Yafang Zhou
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Yacen Hu
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Qiying Sun
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Nina Xie
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| |
Collapse
|
22
|
Emerging Role of mTOR Signaling-Related miRNAs in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6141902. [PMID: 30305865 PMCID: PMC6165581 DOI: 10.1155/2018/6141902] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Mechanistic/mammalian target of rapamycin (mTOR), an atypical serine/threonine kinase of the phosphoinositide 3-kinase- (PI3K-) related kinase family, elicits a vital role in diverse cellular processes, including cellular growth, proliferation, survival, protein synthesis, autophagy, and metabolism. In the cardiovascular system, the mTOR signaling pathway integrates both intracellular and extracellular signals and serves as a central regulator of both physiological and pathological processes. MicroRNAs (miRs), a class of short noncoding RNA, are an emerging intricate posttranscriptional modulator of critical gene expression for the development and maintenance of homeostasis across a wide array of tissues, including the cardiovascular system. Over the last decade, numerous studies have revealed an interplay between miRNAs and the mTOR signaling circuit in the different cardiovascular pathophysiology, like myocardial infarction, hypertrophy, fibrosis, heart failure, arrhythmia, inflammation, and atherosclerosis. In this review, we provide a comprehensive state of the current knowledge regarding the mechanisms of interactions between the mTOR signaling pathway and miRs. We have also highlighted the latest advances on mTOR-targeted therapy in clinical trials and the new perspective therapeutic strategies with mTOR-targeting miRs in cardiovascular diseases.
Collapse
|
23
|
Kanda Y, Kawaguchi T, Osaki M, Onuma K, Ochiya T, Kitagawa T, Okada F. Fascin protein stabilization by miR-146a implicated in the process of a chronic inflammation-related colon carcinogenesis model. Inflamm Res 2018; 67:839-846. [PMID: 30056535 DOI: 10.1007/s00011-018-1175-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE In sporadic colon tumors, multistep process of well-known genetic alterations accelerates carcinogenesis; however, this does not appear to be the case in inflammation-related ones. We previously established a model of inflammation-related colon carcinogenesis using human colonic adenoma cells, and identified fascin as a driver gene of this process. We analyzed the microRNAs involved in the stable fascin expression in colon adenocarcinoma cells. MATERIALS AND METHODS miRNA microarray analysis was performed using FPCK-1-1 adenoma cells and its-derived FPCKpP1-4 adenocarcinoma cells through chronic inflammation. To assess the involvement of miRNA in the inflammation-related carcinogenesis, sphere-forming ability, expression of colon cancer stemness markers, and stability of fascin protein via the proteasome using tough decoy RNA technique. RESULTS We found that 17 miRNAs including miR-146a were upregulated and 16 miRNAs were downregulated in FPCKpP1-4 adenocarcinoma cells. We revealed that miR-146a in the adenocarcinoma cells brought about acquisition of sphere formation, cancer stemness, and inhibition of proteasomal degradation of the fascin protein. CONCLUSIONS We found that stable fascin expression is brought about via the inhibition of proteasome degradation by miR-146a in the process of a chronic inflammation-related colon carcinogenesis.
Collapse
Affiliation(s)
- Yusuke Kanda
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, 86 Nishicho, Yonago, 683-8503, Japan
| | - Tokuichi Kawaguchi
- Japanese Foundation for Cancer Research, Cancer Institute, Tokyo, 135-8550, Japan
| | - Mitsuhiko Osaki
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, 86 Nishicho, Yonago, 683-8503, Japan
- Chromosome Engineering Research Center, Tottori University, Yonago, 683-8503, Japan
| | - Kunishige Onuma
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, 86 Nishicho, Yonago, 683-8503, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tomoyuki Kitagawa
- Japanese Foundation for Cancer Research, Cancer Institute, Tokyo, 135-8550, Japan
| | - Futoshi Okada
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, 86 Nishicho, Yonago, 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, Yonago, 683-8503, Japan.
| |
Collapse
|
24
|
Deciphering the Far-Reaching Functions of Non-coding RNA in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0408-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
25
|
Wang F, Dai CQ, Zhang LR, Bing C, Qin J, Liu YF. Downregulation of Lgr6 inhibits proliferation and invasion and increases apoptosis in human colorectal cancer. Int J Mol Med 2018; 42:625-632. [PMID: 29693156 DOI: 10.3892/ijmm.2018.3633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/30/2018] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to analyze the role of leucine‑rich repeat‑containing G‑protein coupled receptor 6 (Lgr6) in the proliferation and invasion of colorectal cancer (CRC) cells, and to investigate its possible mechanisms. The expression of Lgr6 in CRC tissues was observed by real time‑quantitative polymerase chain reaction and western blotting. Then cell viability, apoptosis and cell invasion was measured by MTT, flow cytometry or Matrigel‑Transwell system, respectively in CRC cells after transfected with Lgr6 siRNA or Lgr6 vector. Furthermore, the expression of apoptosis‑associated protein and PI3K/AKT signaling (phosphorylated‑PI3K, phosphorylated‑AKT, t‑PI3K, t‑AKT) were measured by real‑time PCR/or western blot analysis. The results demonstrated that the level of Lgr6 was higher in CRC tissues than that in adjacent tissues, and Lgr6 overexpression increased CRC proliferation, and invasion of CRC cells in vitro. Notably, suppressing the expression of Lgr6 in CRC cells increased the expression of B‑cell lymphoma-2 (Bcl‑2)‑associated X protein and caspase‑3, but decreased the expression of Bcl‑2 at the mRNA and protein levels. Lgr6 also had the ability to regulate the phosphoinositide 3‑kinase/AKT signaling pathway. It was concluded that Lgr6 has a tumor‑promoting role in the development of CRC, and may serve as a potential diagnostic and prognostic biomarker for the disease.
Collapse
Affiliation(s)
- Fei Wang
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Chun-Qian Dai
- Department of General Surgery, Rudong No. 2 People's Hospital, Rudong, Jiangsu 226400, P.R. China
| | - Li-Rong Zhang
- Department of General Surgery, Rudong No. 2 People's Hospital, Rudong, Jiangsu 226400, P.R. China
| | - Cao Bing
- Department of General Surgery, Rudong No. 2 People's Hospital, Rudong, Jiangsu 226400, P.R. China
| | - Jun Qin
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Yi-Fei Liu
- Department of Pathology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
26
|
Sestrin 2 suppresses cells proliferation through AMPK/mTORC1 pathway activation in colorectal cancer. Oncotarget 2018; 8:49318-49328. [PMID: 28525387 PMCID: PMC5564770 DOI: 10.18632/oncotarget.17595] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/17/2017] [Indexed: 12/25/2022] Open
Abstract
Sestrin 2 is a conserved antioxidant protein that reduces reactive oxygen species (ROS) and inhibits mammalian target of rapamycin complex 1 (mTORC1). We previously showed that sestrin 2 is abnormally decreased in colorectal cancer (CRC). To elucidate the molecular mechanism behind the potential contribution of sestrin 2 to CRC, we used a lentiviral expression vector system to determine the effects of sestrin 2 overexpression on human CRC cells. We found that sestrin 2 overexpression decreased ROS production, inhibited cell growth, and stimulated apoptosis in two CRC cell lines. In parallel, expression of the proliferation marker PCNA was decreased, proapoptotic caspase 3, 7, and 9 levels were increased, and expression of the anti-apoptotic protein survivin was reduced. Sestrin 2 overexpression also activated the adenosine monophosphate-activated protein kinase (AMPK) pathway, and suppressed mTORC1 signaling. Treating CRC cells with compound C, an AMPK inhibitor, reversed or attenuated changes in proliferation, apoptosis, and signaling proteins of the AMPK/mTORC1 axis. In a xenograft mouse model, CRC growth was attenuated by sestrin 2 overexpression. These results suggest that sestrin 2 suppresses CRC cell growth through activation of the AMPK/mTORC1 pathway and induction of apoptosis, and could be a novel pharmacological target for the treatment of CRC.
Collapse
|
27
|
Marshall J, Zhou XZ, Chen G, Yang SQ, Li Y, Wang Y, Zhang ZQ, Jiang Q, Birnbaumer L, Cao C. Antidepression action of BDNF requires and is mimicked by Gαi1/3 expression in the hippocampus. Proc Natl Acad Sci U S A 2018; 115:E3549-E3558. [PMID: 29507199 PMCID: PMC5899481 DOI: 10.1073/pnas.1722493115] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Stress-related alterations in brain-derived neurotrophic factor (BDNF) expression, a neurotrophin that plays a key role in synaptic plasticity, are believed to contribute to the pathophysiology of depression. Here, we show that in a chronic mild stress (CMS) model of depression the Gαi1 and Gαi3 subunits of heterotrimeric G proteins are down-regulated in the hippocampus, a key limbic structure associated with major depressive disorder. We provide evidence that Gαi1 and Gαi3 (Gαi1/3) are required for the activation of TrkB downstream signaling pathways. In mouse embryonic fibroblasts (MEFs) and CNS neurons, Gαi1/3 knockdown inhibited BDNF-induced tropomyosin-related kinase B (TrkB) endocytosis, adaptor protein activation, and Akt-mTORC1 and Erk-MAPK signaling. Functional studies show that Gαi1 and Gαi3 knockdown decreases the number of dendrites and dendritic spines in hippocampal neurons. In vivo, hippocampal Gαi1/3 knockdown after bilateral microinjection of lentiviral constructs containing Gαi1 and Gαi3 shRNA elicited depressive behaviors. Critically, exogenous expression of Gαi3 in the hippocampus reversed depressive behaviors in CMS mice. Similar results were observed in Gαi1/Gαi3 double-knockout mice, which exhibited severe depressive behaviors. These results demonstrate that heterotrimeric Gαi1 and Gαi3 proteins are essential for TrkB signaling and that disruption of Gαi1 or Gαi3 function could contribute to depressive behaviors.
Collapse
MESH Headings
- Animals
- Brain-Derived Neurotrophic Factor/metabolism
- Dendrites/metabolism
- Dendrites/pathology
- Dendritic Spines/metabolism
- Dendritic Spines/pathology
- Depression/metabolism
- Depression/pathology
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/pathology
- Down-Regulation
- Female
- GTP-Binding Protein alpha Subunit, Gi2/biosynthesis
- GTP-Binding Protein alpha Subunit, Gi2/genetics
- GTP-Binding Protein alpha Subunit, Gi2/metabolism
- GTP-Binding Protein alpha Subunits, Gi-Go/biosynthesis
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Hippocampus/metabolism
- Mice
- Mice, Knockout
- Neurons/metabolism
- Neurons/pathology
- Signal Transduction/drug effects
- Stress, Physiological/physiology
Collapse
Affiliation(s)
- John Marshall
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912;
| | - Xiao-Zhong Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004 Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu, China
| | - Su-Qing Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
| | - Ya Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
| | - Yin Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
| | - Zhi-Qing Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
| | - Qin Jiang
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, 210029 Nanjing, China
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709;
- School of Medical Sciences, Institute of Biomedical Research, Catholic University of Argentina, C1107AAZ Buenos Aires, Argentina
| | - Cong Cao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, Soochow University, Suzhou 215000, China;
- Institute of Neuroscience, Soochow University, Suzhou 215000, China
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, 210029 Nanjing, China
- North District, The Municipal Hospital of Suzhou, Suzhou 215001, China
| |
Collapse
|
28
|
Chen MB, Liu YY, Xing ZY, Zhang ZQ, Jiang Q, Lu PH, Cao C. Itraconazole-Induced Inhibition on Human Esophageal Cancer Cell Growth Requires AMPK Activation. Mol Cancer Ther 2018; 17:1229-1239. [PMID: 29592879 DOI: 10.1158/1535-7163.mct-17-1094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/11/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
We here evaluated the antiesophageal cancer cell activity by the antifungal drug itraconazole. Our results show that μg/mL concentrations of itraconazole potently inhibited survival and proliferation of established (TE-1 and Eca-109) and primary human esophageal cancer cells. Itraconazole activated AMPK signaling, which was required for subsequent esophageal cancer cell death. Pharmacologic AMPK inhibition, AMPKα1 shRNA, or dominant negative mutation (T172A) almost completely abolished itraconazole-induced cytotoxicity against esophageal cancer cells. Significantly, itraconazole induced AMPK-dependent autophagic cell death (but not apoptosis) in esophageal cancer cells. Furthermore, AMPK activation by itraconazole induced multiple receptor tyrosine kinases (RTKs: EGFR, PDGFRα, and PDGFRβ), lysosomal translocation, and degradation to inhibit downstream Akt activation. In vivo, itraconazole oral gavage potently inhibited Eca-109 tumor growth in SCID mice. It was yet ineffective against AMPKα1 shRNA-expressing Eca-109 tumors. The in vivo growth of the primary human esophageal cancer cells was also significantly inhibited by itraconazole administration. AMPK activation, RTK degradation, and Akt inhibition were observed in itraconazole-treated tumors. Together, itraconazole inhibits esophageal cancer cell growth via activating AMPK signaling. Mol Cancer Ther; 17(6); 1229-39. ©2018 AACR.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Radiotherapy & Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Zhao-Yu Xing
- The Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhi-Qing Zhang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China.
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Suzhou, China. .,The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.,The Municipal Hospital of Suzhou, North District, Suzhou, China
| |
Collapse
|
29
|
Chen MB, Liu YY, Cheng LB, Lu JW, Zeng P, Lu PH. AMPKα phosphatase Ppm1E upregulation in human gastric cancer is required for cell proliferation. Oncotarget 2018; 8:31288-31296. [PMID: 28423719 PMCID: PMC5458207 DOI: 10.18632/oncotarget.16126] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/15/2017] [Indexed: 12/21/2022] Open
Abstract
Activation of AMP-activated protein kinase (AMPK) is a valuable anti-cancer strategy. In the current study, we tested expression and potential function of Ca2+/calmodulin-dependent protein kinase phosphatase (Ppm1E), an AMPKα phosphatase, in human gastric cancers. Ppm1E expression was elevated in human gastric cancer tissues (vs. normal tissues), which was correlated with AMPK (p-AMPKα, Thr-172) dephosphorylation and mTOR complex 1 (mTORC1) activation. Ppm1E upregulation, AMPK inhibition and mTORC1 activation were also observed in human gastric cancer cell lines (AGS, HGC-27, and SNU601). Intriguingly, Ppm1E knockdown by shRNA induced AMPK activation, mTORC1 inactivation, and proliferation inhibition in AGS cells. On the other hand, forced over-expression of Ppm1E induced further AMPK inhibition and mTORC1 activation to enhance AGS cell proliferation. Remarkably, microRNA-135b-5p (“miR-135b-5p”), an anti-Ppm1E microRNA, was downregulated in both human gastric cancer tissues and cells. Reversely, miR-135b-5p exogenous expression caused Ppm1E depletion, AMPK activation, and AGC cell proliferation inhibition. Together, Ppm1E upregulation in human gastric cancer is important for cell proliferation, possible via regulating AMPK-mTOR signaling.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yuan-Yuan Liu
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Li-Bo Cheng
- Department of Ophthalmology, Wuxi Second Hospital, Nanjing Medical University, Wu'xi, China
| | - Jian-Wei Lu
- Department of Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Ping Zeng
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Pei-Hua Lu
- Department of Radiotherapy and Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
30
|
Cai S, Li Y, Bai JY, Zhang ZQ, Wang Y, Qiao YB, Zhou XZ, Yang B, Tian Y, Cao C. Gαi3 nuclear translocation causes irradiation resistance in human glioma cells. Oncotarget 2018; 8:35061-35068. [PMID: 28456783 PMCID: PMC5471034 DOI: 10.18632/oncotarget.17043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/30/2017] [Indexed: 12/27/2022] Open
Abstract
We have previously shown that Gαi3 is elevated in human glioma, mediating Akt activation and cancer cell proliferation. Here, we imply that Gαi3 could also be important for irradiation resistance. In A172 human glioma cells, Gαi3 knockdown (by targeted shRNAs) or dominant-negative mutation significantly potentiated irradiation-induced cell apoptosis. Reversely, forced over-expression of wild-type or constitutively-active Gαi3 inhibited irradiation-induced A172 cell apoptosis. Irradiation in A172 cells induced Gαi3 translocation to cell nuclei and association with local protein DNA-dependent protein kinase (DNA-PK) catalytic subunit. This association was important for DNA damage repair. Gαi3 knockdown, depletion (using Gαi3 knockout MEFs) or dominant-negative mutation potentiated irradiation-induced DNA damages. On the other hand, expression of the constitutively-active Gαi3 in A172 cells inhibited DNA damage by irradiation. Together, these results indicate a novel function of Gαi3 in irradiation-resistance in human glioma cells.
Collapse
Affiliation(s)
- Shang Cai
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ya Li
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-Yu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi-Qing Zhang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yin Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yin-Biao Qiao
- Department of Surgery, The Third Hospital affiliated to Soochow University
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bo Yang
- Department of Surgery, The Third Hospital affiliated to Soochow University
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Chen MB, Ji XZ, Liu YY, Zeng P, Xu XY, Ma R, Guo ZD, Lu JW, Feng JF. Ulk1 over-expression in human gastric cancer is correlated with patients' T classification and cancer relapse. Oncotarget 2018; 8:33704-33712. [PMID: 28410240 PMCID: PMC5464904 DOI: 10.18632/oncotarget.16734] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/04/2017] [Indexed: 12/26/2022] Open
Abstract
Ulk1 is a key autophagy protein. Here, we tested expression and potential function of Ulk1 in human gastric cancer. Ulk1 mRNA and protein were significantly elevated in multiple fresh human gastric cancer tissues. Its level was relatively low in surrounding normal epithelial tissues. Ulk1 over-expression was also observed in several gastric cancer cell lines (AGS, HGC-27, and SNU601). Remarkably, Ulk1 knockdown by targeted-shRNA inhibited AGS gastric cancer cell survival and proliferation. On the other hand, exogenous Ulk1 over-expression could further promote AGS cell survival and proliferation. Immunohistochemistry (IHC) staining assay of 145 paraffin-embedded gastric cancer tissues showed that Ulk1 was over-expressed in majority (114 out of 145) of gastric cancer tissues. Importantly, high Ulk1 expression in gastric cancer was correlated with patients' T classification and cancer relapse. Together, we demonstrate that Ulk1 over-expression in human gastric cancer is pro-survival. Its over-expression is associated with patients' T classification and cancer relapse.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Radiotherapy & Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu Province, China
| | - Xiao-Zhi Ji
- Departments of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Yuan-Yuan Liu
- Department of Radiotherapy & Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu Province, China
| | - Ping Zeng
- Department of Radiotherapy & Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu Province, China
| | - Xin-Yu Xu
- Departments of Pathology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu Province, China
| | - Rong Ma
- Clinical Cancer Research Center, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zheng-Dong Guo
- Departments of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Jian-Wei Lu
- Departments of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu Province, China.,Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Ji-Feng Feng
- Departments of Medical Oncology, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Jiangsu Province Institute of Cancer, Nanjing, Jiangsu Province, China
| |
Collapse
|
32
|
Li ZW, Zhu YR, Zhou XZ, Zhuo BB, Wang XD. microRNA-135b expression silences Ppm1e to provoke AMPK activation and inhibit osteoblastoma cell proliferation. Oncotarget 2018; 8:26424-26433. [PMID: 28460435 PMCID: PMC5432269 DOI: 10.18632/oncotarget.15477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 02/08/2017] [Indexed: 12/25/2022] Open
Abstract
Forced-activation of AMP-activated protein kinase (AMPK) can possibly inhibit osteoblastoma cells. Here, we aim to provoke AMPK activation via microRNA silencing its phosphatase Ppm1e (protein phosphatase Mg2+/Mn2+-dependent 1e). We showed that microRNA-135b-5p (“miR-135b-5p”), the anti-Ppm1e microRNA, was significantly downregulated in human osteoblastoma tissues. It was correlated with Ppm1e upregulation and AMPKα1 de-phosphorylation. Forced-expression of miR-135b-5p in human osteoblastoma cells (MG-63 and U2OS lines) silenced Ppm1e, and induced a profound AMPKα1 phosphorylation (at Thr-172). Osteoblastoma cell proliferation was inhibited after miR-135b-5p expression. Intriguingly, Ppm1e shRNA knockdown similarly induced AMPKα1 phosphorylation, causing osteoblastoma cell proliferation. Reversely, AMPKα1 shRNA knockdown or dominant negative mutation almost abolished miR-135b-5p's actions in osteoblastoma cells. Further in vivo studies demonstrated that U2OS tumor growth in mice was dramatically inhibited after expressing miR-135b-5p or Ppm1e shRNA. Together, our results suggest that miR-135b-induced Ppm1e silence induces AMPK activation to inhibit osteoblastoma cell proliferation.
Collapse
Affiliation(s)
- Zheng-Wei Li
- The Center of Diagnosis and Treatment for Children's Bone Diseases, The Children's Hospital Affiliated to Soochow University, Suzhou, China
| | - Yun-Rong Zhu
- Department of Orthopedics, The Affiliated Jiangyin Hospital of Medical College of Southeast University, Jiangyin City, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Orthopedics, The First People's Hospital of SuQian, SuQian, China
| | - Bao-Biao Zhuo
- The Center of Diagnosis and Treatment for Children's Bone Diseases, The Children's Hospital Affiliated to Soochow University, Suzhou, China
| | - Xiao-Dong Wang
- The Center of Diagnosis and Treatment for Children's Bone Diseases, The Children's Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
33
|
Cheng L, Liu YY, Lu PH, Peng Y, Yuan Q, Gu XS, Jin Y, Chen MB, Bai XM. Identification of DNA-PKcs as a primary resistance factor of TIC10 in hepatocellular carcinoma cells. Oncotarget 2018; 8:28385-28394. [PMID: 28415690 PMCID: PMC5438657 DOI: 10.18632/oncotarget.16073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/27/2017] [Indexed: 01/07/2023] Open
Abstract
The current study tested the anti-hepatocellular carcinoma (HCC) cell activity of TIC10, a first-in-class small-molecule tumor necrosis (TNF)-related apoptosis-inducing ligand (TRAIL) inducer. TIC10 exerted potent anti-proliferative and pro-apoptotic actions in primary and established human HCC cells. TIC10 blocked Akt-Erk activation, leading to Foxo3a nuclear translocation, as well as TRAIL and death receptor-5 (DR5) transcription in HCC cells. We propose that DNA-PKcs is a major resistance factor of TIC10 possibly via inhibiting Foxo3a nuclear translocation. DNA-PKcs inhibition, knockdown or mutation facilitated TIC10-induced Foxo3a nuclear translocation, TRAIL/DR5 expression and cell apoptosis. Reversely, exogenous DNA-PKcs over-expression inhibited above actions by TIC10. In vivo, oral administration of TIC10 significantly inhibited HepG2 tumor growth in nude mice, which was further potentiated with Nu7026 co-administration. Thus, TIC10 shows promising anti-HCC activity, alone or together with DNA-PKcs inhibitors.
Collapse
Affiliation(s)
- Long Cheng
- Department of Interventional Radiology, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yuan-Yuan Liu
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Wuhan, China
| | - Qiang Yuan
- Department of Interventional Radiology, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Xin-Shi Gu
- Department of Interventional Radiology, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yong Jin
- Department of Interventional Radiology, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Min-Bin Chen
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Xu-Ming Bai
- Department of Interventional Radiology, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
34
|
Fan JB, Liu W, Zhu XH, Yi H, Cui SY, Zhao JN, Cui ZM. microRNA-25 targets PKCζ and protects osteoblastic cells from dexamethasone via activating AMPK signaling. Oncotarget 2018; 8:3226-3236. [PMID: 27911275 PMCID: PMC5356877 DOI: 10.18632/oncotarget.13698] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
AMP-activated protein kinase (AMPK) activation could protect osteoblasts from dexamethasone (Dex). This study aims to provoke AMPK activation via microRNA downregulation of its negative regulator protein kinase C ζ (PKCζ). Results show that microRNA-25-5p (miR-25-5p) targets PKCζ's 3’ untranslated regions (UTRs). Forced-expression of miR-25 downregulated PKCζ and activated AMPK in human osteoblastic cells (OB-6 and hFOB1.19 lines), which thereafter protected cells from Dex. Reversely, expression of antagomiR-25, the miR-25 inhibitor, upregulated PKCζ and inhibited AMPK activation, exacerbating Dex damages. Notably, PKCζ shRNA knockdown similarly activated AMPK and protected osteoblastic cells from Dex. AMPK activation was required for miR-25-induced osteoblastic cell protection. AMPKα shRNA or dominant negative mutation almost completely blocked miR-25-induced cytoprotection against Dex. Further studies showed that miR-25 expression increased NADPH activity and suppressed Dex-induced oxidative stress in osteoblastic cells. Such effects by miR-25 were abolished with AMPKα knockdown or mutation. Significantly, miR-25-5p level was increased in patients’ necrotic femoral head tissues, which was correlated with PKCζ downregulation and AMPK hyper-activation. These results suggest that miR-25-5p targets PKCζ and protects osteoblastic cells from Dex possibly via activating AMPK signaling.
Collapse
Affiliation(s)
- Jian-Bo Fan
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China.,Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing 210008, Jiangsu, PR China
| | - Wei Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Xin-Hui Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Hong Yi
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Sheng-Yu Cui
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Jian-Ning Zhao
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing 210008, Jiangsu, PR China
| | - Zhi-Ming Cui
- Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| |
Collapse
|
35
|
Lu XS, Qiao YB, Li Y, Yang B, Chen MB, Xing CG. Preclinical study of cinobufagin as a promising anti-colorectal cancer agent. Oncotarget 2018; 8:988-998. [PMID: 27894091 PMCID: PMC5352212 DOI: 10.18632/oncotarget.13519] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 10/28/2016] [Indexed: 12/21/2022] Open
Abstract
Here, we assessed the anti-colorectal cancer (CRC) cell activity of cinobufagin (CBG). We found that CBG exerted potent cytotoxic and anti-proliferative activity against CRC lines (HCT-116 and HT-29) and primary human CRC cells. Meanwhile, it activated apoptosis, and disrupted cell-cycle progression in the cells. At the signaling level, CBG treatment in CRC cells provoked endoplasmic reticulum stress (ER stress), the latter was evidenced by caspase-12 activation, CHOP expression, as well as PERK and IRE1 phosphorylations. Contrarily, the ER stress inhibitor salubrinal, the caspase-12 inhibitor and CHOP shRNA remarkably attenuated CBG-induced CRC cell death and apoptosis. Further, CBG in-activated mammalian target or rapamycin complex 1 (mTORC1), which appeared responsible for proliferation inhibition in CRC cells. Introduction of a constitutively-active S6K1 (“ca-S6K1”) restored proliferation of CBG-treated CRC cells. Finally, CBG intraperitoneal injection suppressed HCT-116 xenograft tumor growth in the nude mice. CHOP upregulation and mTORC1 in-activation were also noticed in CBG-treated HCT-116 tumors. The results of this preclinical study suggest that CBG could be tested as promising anti-CRC agent.
Collapse
Affiliation(s)
- Xing-Sheng Lu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of General Surgery, Suzhou Municipal Hospital, Suzhou, China
| | - Yin-Biao Qiao
- Department of Hepatobiliary Surgery, The Third Hospital Affiliated to Soochow University, Changzhou City, Jiangsu, China
| | - Ya Li
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bo Yang
- Department of Hepatobiliary Surgery, The Third Hospital Affiliated to Soochow University, Changzhou City, Jiangsu, China
| | - Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Chun-Gen Xing
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
Fan JB, Ruan JW, Liu W, Zhu LQ, Zhu XH, Yi H, Cui SY, Zhao JN, Cui ZM. miR-135b expression downregulates Ppm1e to activate AMPK signaling and protect osteoblastic cells from dexamethasone. Oncotarget 2018; 7:70613-70622. [PMID: 27661114 PMCID: PMC5342578 DOI: 10.18632/oncotarget.12138] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/06/2016] [Indexed: 12/25/2022] Open
Abstract
Activation of AMP-activated protein kinase (AMPK) could potently protect osteoblasts/osteoblastic cells from dexamethasone (Dex). We aim to induce AMPK activation via microRNA ("miRNA") downregulation of its phosphatase Ppm1e. We discovered that microRNA-135b ("miR-135b") targets the 3' untranslated regions (UTRs) of Ppm1e. In human osteoblasticOB-6 cells and hFOB1.19 cells, forced-expression of miR-135b downregulated Ppm1e and activated AMPK signaling. miR-135b also protected osteoblastic cells from Dex. shRNA-induced knockdown of Ppm1e similarly activated AMPK and inhibited Dex-induced damages. Intriguingly, in the Ppm1e-silenced osteoblastic cells, miR-135b expression failed to offer further cytoprotection against Dex. Notably, AMPK knockdown (via shRNA) or dominant negative mutation abolished miR-135b-induced AMPK activation and cytoprotection against Dex. Molecularly, miR-135b, via activating AMPK, increased nicotinamide adenine dinucleotide phosphate (NADPH) activity and inhibited Dex-induced oxidative stress. At last, we found that miR-135b level was increased in human necrotic femoral head tissues, which was correlated with Ppm1e downregulation and AMPK activation. There results suggest that miR-135b expression downregulates Ppm1e to activate AMPK signaling, which protects osteoblastic cells from Dex.
Collapse
Affiliation(s)
- Jian-Bo Fan
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China.,Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing 210008, Jiangsu, PR China
| | - Jian-Wei Ruan
- The Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Liu
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Lun-Qing Zhu
- The Center of Diagnosis and Treatment for Childrens' Bone Disease, Childrens' Hospital Affiliated to Soochow University, Suzhou 215000, Jiangsu, PR China
| | - Xin-Hui Zhu
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Hong Yi
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Sheng-Yu Cui
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| | - Jian-Ning Zhao
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing 210008, Jiangsu, PR China
| | - Zhi-Ming Cui
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, PR China
| |
Collapse
|
37
|
AntogomiR-451 protects human gastric epithelial cells from ethanol via activating AMPK signaling. Biochem Biophys Res Commun 2018; 497:339-346. [PMID: 29432731 DOI: 10.1016/j.bbrc.2018.02.082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/25/2022]
Abstract
The prevention and treatment efficiency of ethanol-induced gastric epithelial injury are not satisfied. We have previously shown that AMP-activated protein kinase (AMPK) activation exerts a pro-survival function in human gastric epithelial cells (GECs). miroRNA-451 ("miR-451")'s inhibitor, antagomiR-451, can activate AMPK signaling. In the present study, we show that forced-expression of antagomiR-451 via a lentiviral vector depleted miR-451, leading to AMPK activation in established GES-1 cells and primary human GECs. AntagomiR-451 efficiently protected GES-1 cells and primary human GECs from ethanol-induced viability reduction and apoptosis. AMPK activation is required for antagomiR-451-induced GEC protection. AMPKα1 knockdown (by targeted-shRNAs) or knockout (by CRISPR-Cas-9 KO plasmid) blocked antagomiR-451-induced AMPK activation, and GEC protection against ethanol. Further experimental results show that antagomiR-451 significantly attenuated ethanol-induced reactive oxygen species (ROS) production, lipid peroxidation and DNA damage. Collectively, antagomiR-451 protects human GECs from ethanol via activating AMPK signaling.
Collapse
|
38
|
Lu PH, Chen MB, Ji C, Li WT, Wei MX, Wu MH. Aqueous Oldenlandia diffusa extracts inhibits colorectal cancer cells via activating AMP-activated protein kinase signalings. Oncotarget 2018; 7:45889-45900. [PMID: 27322552 PMCID: PMC5216768 DOI: 10.18632/oncotarget.9969] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/03/2016] [Indexed: 11/25/2022] Open
Abstract
Here we evaluated the anti-cancer activity of aqueous Oldenlandia diffusa (OD) extracts (ODE) in colorectal cancer (CRC) cells. We showed that ODE exerted potent anti-proliferative, cytotoxic and pro-apoptotic activities against a panel of established CRC lines (HCT-116, DLD-1, HT-29 and Lovo) and primary (patient-derived) human CRC cells. ODE activated AMP-activated protein kinase (AMPK) signaling, which led to subsequent mTORC1 inhibition and Bcl-2/HIF-1α downregulation in CRC cells. In ODE-treated CRC cells, AMPKα1 formed a complex with p53. This might be important for p53 activation and subsequent cancer cell apoptosis. Inhibition of AMPK signaling, though dominant negative (dn) mutation or shRNA/siRNA knockdown of AMPKα1 attenuated ODE-exerted CRC cytotoxicity. In vivo, i.p. administration of ODE inhibited HCT-116 xenograft tumor growth in SCID mice. In addition, AMPK activation, mTORC1 inhibition and p53 activation were observed in ODE-treated HCT-116 xenograft tumors. These results suggest that ODE inhibits CRC cells in vitro and in vivo, possibly via activation of AMPK-dependent signalings.
Collapse
Affiliation(s)
- Pei-Hua Lu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, 215300, Jiangsu, China
| | - Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Wen-Ting Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mu-Xin Wei
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Mian-Hua Wu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
39
|
Wang P, Liu XM, Ding L, Zhang XJ, Ma ZL. mTOR signaling-related MicroRNAs and Cancer involvement. J Cancer 2018; 9:667-673. [PMID: 29556324 PMCID: PMC5858488 DOI: 10.7150/jca.22119] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/05/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of single-stranded RNAs, 18-23 nucleotides in length that regulate gene expression at the post-transcriptional level. Dysregulation of miRNAs has been closely associated with the development of cancer. In the process of tumorigenesis, mammalian target of rapamycin (mTOR) plays important roles, and the mTOR signaling pathway is aberrant in various types of human cancers, including non-small cell lung cancer (NSCLC), breast cancer, prostate cancer, as well as others. However, the relationship between miRNAs and the mTOR signaling pathway is indistinct. Herein, we not only summarize the progress of miRNAs and the mTOR signaling pathway in cancers, but also highlight their role in the diagnosis and treatment in the clinic.
Collapse
Affiliation(s)
- Ping Wang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xiao-Min Liu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China.,School of Environmental Science and Engineering, Shanghai University, Shanghai, 200444, China
| | - Lei Ding
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xin-Ju Zhang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhong-Liang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
40
|
Zeng R, Zhang R, Song X, Ni L, Lai Z, Liu C, Ye W. The long non-coding RNA MALAT1 activates Nrf2 signaling to protect human umbilical vein endothelial cells from hydrogen peroxide. Biochem Biophys Res Commun 2017; 495:2532-2538. [PMID: 29274336 DOI: 10.1016/j.bbrc.2017.12.105] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022]
Abstract
The potential effect of the long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) against hydrogen peroxide (H2O2)-induced oxidative injury in endothelial cells was tested. We show that forced-expression of MALAT1 using a lentiviral vector ("LV-MALAT1") significantly attenuated H2O2-induced death and apoptosis of human umbilical vein endothelial cells (HUVECs). Conversely, knocking down of MALAT1 by targeted siRNA exacerbated H2O2-induced HUVEC injury. For the mechanism study, we show that LV-MALAT1 induced Keap1 downregulation, leading to nuclear-factor-E2-related factor 2 (Nrf2) stabilization and activation. Critically, Nrf2 shRNA almost completely abolished LV-MALAT1-mediated HUVEC protection against H2O2. Significantly, H2O2-induced oxidative stress, lipid peroxidation and DNA damages in HUVECs were attenuated by LV-MALAT1, but were intensified with MALAT1 siRNA. In summary, we identified a novel signaling axis involving MALAT1, Keap1 and Nrf2, which in turn protects HUVECs from oxidative injury.
Collapse
Affiliation(s)
- Rong Zeng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rui Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xitao Song
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Leng Ni
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhichao Lai
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Changwei Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Ye
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
41
|
Weng Y, Lin J, Liu H, Wu H, Yan Z, Zhao J. AMPK activation by Tanshinone IIA protects neuronal cells from oxygen-glucose deprivation. Oncotarget 2017; 9:4511-4521. [PMID: 29435120 PMCID: PMC5796991 DOI: 10.18632/oncotarget.23391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 12/25/2022] Open
Abstract
The current study tested the potential neuroprotective function of Tanshinone IIA (ThIIA) in neuronal cells with oxygen-glucose deprivation (ODG) and re-oxygenation (OGDR). In SH-SY5Y neuronal cells and primary murine cortical neurons, ThIIA pre-treatment attenuated OGDR-induced viability reduction and apoptosis. Further, OGDR-induced mitochondrial depolarization, reactive oxygen species production, lipid peroxidation and DNA damages in neuronal cells were significantly attenuated by ThIIA. ThIIA activated AMP-activated protein kinase (AMPK) signaling, which was essential for neuroprotection against OGDR. AMPKα1 knockdown or complete knockout in SH-SY5Y cells abolished ThIIA-induced AMPK activation and neuroprotection against OGDR. Further studies found that ThIIA up-regulated microRNA-135b to downregulate the AMPK phosphatase Ppm1e. Notably, knockdown of Ppm1e by targeted shRNA or forced microRNA-135b expression also activated AMPK and protected SH-SY5Y cells from OGDR. Together, AMPK activation by ThIIA protects neuronal cells from OGDR. microRNA-135b-mediated silence of Ppm1e could be the key mechanism of AMPK activation by ThIIA.
Collapse
Affiliation(s)
- Yingfeng Weng
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jixian Lin
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Liu
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Wu
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhimin Yan
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Zhao
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Chen MB, Zhou ZT, Yang L, Wei MX, Tang M, Ruan TY, Xu JY, Zhou XZ, Chen G, Lu PH. KU-0060648 inhibits hepatocellular carcinoma cells through DNA-PKcs-dependent and DNA-PKcs-independent mechanisms. Oncotarget 2017; 7:17047-59. [PMID: 26933997 PMCID: PMC4941370 DOI: 10.18632/oncotarget.7742] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023] Open
Abstract
Here we tested anti-tumor activity of KU-0060648 in preclinical hepatocellular carcinoma (HCC) models. Our results demonstrated that KU-0060648 was anti-proliferative and pro-apoptotic in established (HepG2, Huh-7 and KYN-2 lines) and primary human HCC cells, but was non-cytotoxic to non-cancerous HL-7702 hepatocytes. DNA-PKcs (DNA-activated protein kinase catalytic subunit) is an important but not exclusive target of KU-0060648. DNA-PKcs knockdown or dominant negative mutation inhibited HCC cell proliferation. On the other hand, overexpression of wild-type DNA-PKcs enhanced HepG2 cell proliferation. Importantly, KU-0060648 was still cytotoxic to DNA-PKcs-silenced or -mutated HepG2 cells, although its activity in these cells was relatively weak. Further studies showed that KU-0060648 inhibited PI3K-AKT-mTOR activation, independent of DNA-PKcs. Introduction of constitutively-active AKT1 (CA-AKT1) restored AKT-mTOR activation after KU-0060648 treatment in HepG2 cells, and alleviated subsequent cytotoxicity. In vivo, intraperitoneal (i.p.) injection of KU-0060648 significantly inhibited HepG2 xenograft growth in nude mice. AKT-mTOR activation was also inhibited in xenografted tumors. Finally, we showed that DNA-PKcs expression was significantly upregulated in human HCC tissues. Yet miRNA-101, an anti-DNA-PKcs miRNA, was downregulated. Over-expression of miR-101 in HepG2 cells inhibited DNA-PKcs expression and cell proliferation. Together, these results indicate that KU-0060648 inhibits HCC cells through DNA-PKcs-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan 215300, China
| | - Zhen-Tao Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lan Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Mu-Xin Wei
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Min Tang
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan 215300, China
| | - Ting-Yan Ruan
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Jun-Ying Xu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Xiao-Zhong Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Gang Chen
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| |
Collapse
|
43
|
Liu HX, Xu MQ, Li SP, Tian S, Guo MX, Qi JY, He CJ, Zhao XS. Jujube leaf green tea extracts inhibits hepatocellular carcinoma cells by activating AMPK. Oncotarget 2017; 8:110566-110575. [PMID: 29299169 PMCID: PMC5746404 DOI: 10.18632/oncotarget.22821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022] Open
Abstract
Here we evaluated the anti-hepatocellular carcinoma activity by the Jujube leaf green tea extracts (JLGTE). We showed that JLGTE exerted anti-proliferative, cytotoxic and pro-apoptotic activities against HepG2 and primary human hepatocellular carcinoma cells. It was however non-cytotoxic to the normal hepatocytes. JLGTE activated AMP-activated protein kinase (AMPK) signaling, which was required for its cytotoxicity against hepatocellular carcinoma cells. Silence of AMPKα1, via targeted short hairpin RNAs or CRISPR-Cas9 genome editing, inhibited JLGTE-induced AMPK activation and HepG2 cell apoptosis. Further, in-activation of AMPK by a dominant negative AMPKα1 (T172A) also alleviated JLGTE's cytotoxicity against HepG2 cells. On the other hand, forced-activation of AMPK by introduction of a constitutively-active AMPKα1 (T172D) mimicked JLGTE's actions and led to HepG2 cell apoptosis. These results suggest that JLGTE inhibits human hepatocellular carcinoma cells possibly via activating AMPK.
Collapse
Affiliation(s)
- H X Liu
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - M Q Xu
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - S P Li
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - S Tian
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - M X Guo
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - J Y Qi
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - C J He
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| | - X S Zhao
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, China
| |
Collapse
|
44
|
Dai C, Zhang X, Xie D, Tang P, Li C, Zuo Y, Jiang B, Xue C. Targeting PP2A activates AMPK signaling to inhibit colorectal cancer cells. Oncotarget 2017; 8:95810-95823. [PMID: 29221169 PMCID: PMC5707063 DOI: 10.18632/oncotarget.21336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022] Open
Abstract
LB-100 is a novel PP2A inhibitor. Its activity in human colorectal cancer (CRC) cells was tested. The in vitro studies demonstrated that LB-100 inhibited survival and proliferation of both established CRC cells (HCT-116 and HT-29 lines) and primary human colon cancer cells. Further, LB-100 activated apoptosis and induced G1-S cell cycle arrest in CRC cells. LB-100 inhibited PP2A activity and activated AMPK signaling in CRC cells. AMPKα1 dominant negative mutation, shRNA-mediated knockdown or complete knockout (by CRISPR/Cas9 method) largely attenuated LB-100-induced AMPK activation and HCT-116 cytotoxicity. Notably, microRNA-17-92-mediated silence of PP2A (regulatory B subunit) also activated AMPK and induced HCT-116 cell death. Such effects were again largely attenuated by AMPKα mutation, silence or complete knockout. In vivo studies showed that intraperitoneal injection of LB-100 inhibited HCT-116 xenograft growth in nude mice. Its anti-tumor activity was largely compromised against HCT-116 tumors-derived from AMPKα1-knockout cells. We conclude that targeting PP2A by LB-100 and microRNA-17-92 activates AMPK signaling to inhibit CRC cells.
Collapse
Affiliation(s)
- Cuiping Dai
- Faculty of Health, Jiangsu Food and Pharmaceutical Science College, Huaian, China
| | - Xuning Zhang
- Huaian Key Laboratory Of Gastrointestinal Cancer, Jiangsu College of Nursing, Huaian, China
| | - Da Xie
- Oncology Department, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peipei Tang
- Huaian Key Laboratory Of Gastrointestinal Cancer, Jiangsu College of Nursing, Huaian, China
| | - Chunmei Li
- Huaian Key Laboratory Of Gastrointestinal Cancer, Jiangsu College of Nursing, Huaian, China
| | - Yi Zuo
- Department of Medicine, Xinglin College, Nantong University, Nantong, China
| | - Baofei Jiang
- Gastrointestinal Surgery, The First People's Hospital of Huaian City, Huaian, China
| | - Caiping Xue
- Huaian Key Laboratory Of Gastrointestinal Cancer, Jiangsu College of Nursing, Huaian, China
| |
Collapse
|
45
|
Lu PH, Chen MB, Liu YY, Wu MH, Li WT, Wei MX, Liu CY, Qin SK. Identification of sphingosine kinase 1 (SphK1) as a primary target of icaritin in hepatocellular carcinoma cells. Oncotarget 2017; 8:22800-22810. [PMID: 28206952 PMCID: PMC5410263 DOI: 10.18632/oncotarget.15205] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/08/2016] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive neoplasm. We aim to explore the anti-HCC activity by a natural prenylflavonoid icaritin. Icaritin was cytotoxic and pro-apoptotic when added to established (HepG2, KYN-2 and Huh-7 lines) and primary human HCC cells. At the signaling level, icaritin inhibited sphingosine kinase 1 (SphK1) activity in HCC cells, which led to pro-apoptotic ceramide production and JNK1 activation. SphK1 inhibition or silence (by shRNA/microRNA) mimicked icaritin-mediated cytotoxicity, and almost nullified icaritin's activity in HepG2 cells. Reversely, exogenous over-expression of SphK1 sensitized icaritin-induced HepG2 cell apoptosis. In vivo, oral administration of icaritin dramatically inhibited HepG2 xenograft growth in SCID mice. Further, SphK1 activity in icaritin-treated tumors was largely inhibited. In summary, icaritin exerts potent anti-HCC activity in vitro and in vivo. SphK1 inhibition could be the primary mechanism of its actions in HCC cells.
Collapse
Affiliation(s)
- Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min-Bin Chen
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yuan-Yuan Liu
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Mian-Hua Wu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Ting Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mu-Xin Wei
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chao-Ying Liu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Shu-Kui Qin
- People's Liberation Army Cancer Center, 81st Hospital of People's Liberation Army, Nanjing, China
| |
Collapse
|
46
|
Zhao Z, Feng L, Wang J, Cheng D, Liu M, Ling M, Xu W, Sun K. NPC-26 kills human colorectal cancer cells via activating AMPK signaling. Oncotarget 2017; 8:18312-18321. [PMID: 28407688 PMCID: PMC5392330 DOI: 10.18632/oncotarget.15436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/11/2017] [Indexed: 12/25/2022] Open
Abstract
NPC-26 is novel mitochondrion-interfering compound. The current study tested its potential effect against colorectal cancer (CRC) cells. We demonstrated that NPC-26 induced potent anti-proliferative and cytotoxic activities against CRC cell lines (HCT-116, DLD-1 and HT-29). Activation of AMP-activated protein kinase (AMPK) signaling mediated NPC-26-induced CRC cell death. AMPKα1 shRNA knockdown or dominant negative mutation abolished NPC-26-induced AMPK activation and subsequent CRC cell death. NPC-26 disrupted mitochondrial function, causing mitochondrial permeability transition pore (mPTP) opening and reactive oxygen species (ROS) production. ROS scavengers (NAC or MnTBAP) and mPTP blockers (cyclosporin A or sanglifehrin A) blocked NPC-26-induced AMPK activation and attenuated CRC cell death. Significantly, intraperitoneal injection of NPC-26 potently inhibited HCT-116 tumor growth in severe combined immuno-deficient (SCID) mice. Yet, its anti-tumor activity was significantly weakened against AMPKα1-silenced HCT-116 tumors. Together, we conclude that NPC-26 kills CRC cells possibly via activating AMPK signaling.
Collapse
Affiliation(s)
- Zhen Zhao
- Clinical Laboratory, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiqin Wang
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Deshan Cheng
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Mei Liu
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Meirong Ling
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| | - Weiping Xu
- Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Keyu Sun
- Emergency Department, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Alamoudi AA, Alnoury A, Gad H. miRNA in tumour metabolism and why could it be the preferred pathway for energy reprograming. Brief Funct Genomics 2017; 17:157-169. [DOI: 10.1093/bfgp/elx023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
48
|
Cao J, Yan XR, Liu T, Han XB, Yu JJ, Liu SH, Wang LB. MicroRNA-552 promotes tumor cell proliferation and migration by directly targeting DACH1 via the Wnt/β-catenin signaling pathway in colorectal cancer. Oncol Lett 2017; 14:3795-3802. [PMID: 28927149 DOI: 10.3892/ol.2017.6600] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 01/31/2017] [Indexed: 12/18/2022] Open
Abstract
The purpose of the present study was to analyze the crucial role of microRNAs (miRNAs/miRs) involved in the proliferation and migration of colorectal cancer (CRC) and to investigate their underlying mechanisms. The present study discusses the expression and function of miR-552 in CRC. The expression level of miR-552 in CRC cells and tissues was observed, and it was suggested that the high expression of miR-552 accelerated the proliferation and migration of CRC cells in vitro. Notably, a result of the present study was that the cell fate determination factor Dachshund family transcription factor 1 (DACH1) was identified as a direct target of miR-552. Suppressing miR-552 expression in CRC cells increased endogenous DACH1 mRNA and protein levels, which was negatively correlated with miR-552. DACH1 performs an important role in the development of a number of neoplasms, and has the ability to regulate the Wnt/β-catenin signaling pathway as a novel predictive and diagnostic biomarker. Accordingly, it was concluded that miR-552 exerted a tumor-promoting role in CRC development by targeting DACH1, which may contribute to the increase in the rates of CRC proliferation and migration. miR-552 may serve as a potential diagnostic and prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Jia Cao
- Department of Medical Laboratory, Ningxia Medical University, Clinical Medicine College, Yinchuan, Ningxia 750004, P.R. China
| | - Xiu-Rui Yan
- Stem Cell Institute, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ting Liu
- Stem Cell Institute, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xue-Bo Han
- Department of Medical Laboratory, Ningxia Medical University, Clinical Medicine College, Yinchuan, Ningxia 750004, P.R. China
| | - Jing-Jing Yu
- Stem Cell Institute, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China.,National Engineering Research Center of Beijing Biochip Technology, Sub-Center in Ningxia, Yinchuan, Ningxia 750004, P.R. China
| | - Shi-Hai Liu
- Central Laboratory, The Affiliated Hospital of Qingdao University, Animal Experimental Center, Qingdao, Shandong 266003, P.R. China
| | - Li-Bin Wang
- Department of Medical Laboratory, Ningxia Medical University, Clinical Medicine College, Yinchuan, Ningxia 750004, P.R. China.,Stem Cell Institute, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China.,National Engineering Research Center of Beijing Biochip Technology, Sub-Center in Ningxia, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
49
|
Zhang S, Zhang Y, Cheng Q, Ma Z, Gong G, Deng Z, Xu K, Wang G, Wei Y, Zou X. Silencing protein kinase C ζ by microRNA-25-5p activates AMPK signaling and inhibits colorectal cancer cell proliferation. Oncotarget 2017; 8:65329-65338. [PMID: 29029434 PMCID: PMC5630334 DOI: 10.18632/oncotarget.18649] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/08/2017] [Indexed: 12/25/2022] Open
Abstract
Developing novel strategies against human colorectal cancer (CRC) cells is needed. Activation of AMP-activated protein kinase (AMPK) could possibly inhibit CRC cells. Protein kinase C ζ (PKCζ) is an AMPK negative regulator. Here we found that PKCζ expression was significantly elevated in human colon cancer tissues and CRC cells. PKCζ upregulation was correlated with AMPK in-activation and mTOR complex 1 (mTORC1) over-activation. Reversely, PKCζ shRNA knockdown activated AMPK signaling and inhibited HT-29 cell proliferation. Significantly, downregulation of microRNA-25-5p (miR-25-5p), a PKCζ-targeting miRNA, could be the cause of PKCζ upregulation. Exogenous expression of miR-25-5p silenced PKCζ to activate AMPK signaling, which inhibited HT-29 cell proliferation. In vivo studies showed that HT-29 xenograft growth in mice was inhibited after expressing PKCζ shRNA or miR-25-5p. Collectively, PKCζ could be a novel oncogenic protein of human CRC. PKCζ silence, by targeted-shRNA or miR-25-5p expression, activates AMPK and inhibits HT-29 cell proliferation.
Collapse
Affiliation(s)
- Shihu Zhang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiyang Zhang
- Digestive Department, Affiliated Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, China
| | - Qing Cheng
- Department of Gynaecology and Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhaoqun Ma
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Guanwen Gong
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengming Deng
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Xu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Gaoyuan Wang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yousong Wei
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoping Zou
- Digestive Department, Affiliated Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, China
| |
Collapse
|
50
|
Micó V, Berninches L, Tapia J, Daimiel L. NutrimiRAging: Micromanaging Nutrient Sensing Pathways through Nutrition to Promote Healthy Aging. Int J Mol Sci 2017; 18:E915. [PMID: 28445443 PMCID: PMC5454828 DOI: 10.3390/ijms18050915] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
Current sociodemographic predictions point to a demographic shift in developed and developing countries that will result in an unprecedented increase of the elderly population. This will be accompanied by an increase in age-related conditions that will strongly impair human health and quality of life. For this reason, aging is a major concern worldwide. Healthy aging depends on a combination of individual genetic factors and external environmental factors. Diet has been proved to be a powerful tool to modulate aging and caloric restriction has emerged as a valuable intervention in this regard. However, many questions about how a controlled caloric restriction intervention affects aging-related processes are still unanswered. Nutrient sensing pathways become deregulated with age and lose effectiveness with age. These pathways are a link between diet and aging. Thus, fully understanding this link is a mandatory step before bringing caloric restriction into practice. MicroRNAs have emerged as important regulators of cellular functions and can be modified by diet. Some microRNAs target genes encoding proteins and enzymes belonging to the nutrient sensing pathways and, therefore, may play key roles in the modulation of the aging process. In this review, we aimed to show the relationship between diet, nutrient sensing pathways and microRNAs in the context of aging.
Collapse
Affiliation(s)
- Víctor Micó
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Laura Berninches
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Javier Tapia
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Lidia Daimiel
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
- Department of Nutrition and Bromatology, CEU San Pablo University, Boadilla del Monte, 28668 Madrid, Spain.
| |
Collapse
|