351
|
Jafarzadeh M, Soltani BM, Soleimani M, Hosseinkhani S. Epigenetically silenced LINC02381 functions as a tumor suppressor by regulating PI3K-Akt signaling pathway. Biochimie 2020; 171-172:63-71. [DOI: 10.1016/j.biochi.2020.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/18/2020] [Indexed: 12/21/2022]
|
352
|
Yousef EH, El-Mesery ME, Habeeb MR, Eissa LA. Polo-like kinase 1 as a promising diagnostic biomarker and potential therapeutic target for hepatocellular carcinoma. Tumour Biol 2020; 42:1010428320914475. [PMID: 32252611 DOI: 10.1177/1010428320914475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular carcinoma is a major cause of cancer mortality worldwide. The outcome of hepatocellular carcinoma depends mainly on its early diagnosis. To date, the performance of traditional biomarkers is unsatisfactory. Polo-like kinase 1 is a serine/threonine kinase that plays essential roles in cell cycle progression and deoxyribonucleic acid damage. Moreover, polo-like kinase 1 knockdown decreases the survival of hepatocellular carcinoma cells; therefore, polo-like kinase 1 is an attractive target for anticancer treatments. Nobiletin, a natural polymethoxy flavonoid, exhibits a potential antiproliferative effect against a wide variety of cancers. This study targets to identify a reliable diagnostic biomarker for hepatocellular carcinoma and provide a potential therapeutic target for its treatment. Polo-like kinase 1 levels were analyzed in 44 hepatocellular carcinoma patients, 33 non-hepatocellular carcinoma liver cirrhosis patients and 15 healthy controls using the enzyme-linked immunosorbent assay method. Receiver operating characteristics curve analysis was used to establish a predictive model for polo-like kinase 1 relative to α-fetoprotein in hepatocellular carcinoma diagnosis. Furthermore, in the in vitro study, gene expressions were assessed by quantitative polymerase chain reaction in two human hepatocellular carcinoma cell lines after treatment with doxorubicin and polo-like kinase 1 inhibitor volasertib (Vola) either alone or in combination with nobiletin. Cell viability was also determined using the crystal violet assay.: Serum polo-like kinase 1 levels in hepatocellular carcinoma patients were significantly higher than liver cirrhosis and control groups (p < 0.0001). Polo-like kinase 1 showed a reasonable sensitivity, specificity, positive predictive value, and negative predictive value in hepatocellular carcinoma diagnosis. Moreover, nobiletin improved inhibition of cell growth induced by Vola and doxorubicin. Regarding reverse transcription polymerase chain reaction results, nobiletin suppressed expressions of polo-like kinase 1 and proliferating cell nuclear antigen and elevated expressions of P53, poly (ADPribose) polymerase 1, and caspase-3. Nobiletin/doxorubicin and nobiletin/Vola showed a significant increase in caspase-3 activity indicating cell apoptosis. Polo-like kinase 1 may be a potential biomarker for hepatocellular carcinoma diagnosis and follow-up during treatment with chemotherapies. In addition, nobiletin synergistically potentiates the doxorubicin and Vola-mediated anticancer effect that may be attributed partly to suppression of polo-like kinase 1 and proliferating cell nuclear antigen expression and enhancement of chemotherapy-induced apoptosis.
Collapse
Affiliation(s)
- Eman H Yousef
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Horus University - Egypt, Damietta, Egypt
| | - Mohamed E El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Maha R Habeeb
- Department of Internal Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Laila A Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
353
|
Ying J, Yang J, Liu Y. LncARSR promotes non-small-cell lung cancer progression via regulating PTEN/Akt. Am J Transl Res 2020; 12:857-866. [PMID: 32269718 PMCID: PMC7137040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/28/2019] [Indexed: 06/11/2023]
Abstract
LncRNAs have been suggested to be key modulators in many biological and pathological processes. LncARSR, a recently identified lncRNA, plays crucial roles in the progression of several cancers. However, the role of lncARSR in NSCLC is uninvestigated. In the present study, it was demonstrated that lncARSR expression was higher in NSCLC tissues than in noncancerous tissues. The expression of lncARSR was higher in four NSCLC cell lines than in a normal lung bronchial epithelial line. Further investigation demonstrated that increased lncARSR expression promoted NSCLC cell migration and growth and induced epithelial-mesenchymal transition in A549 cells. Moreover, ectopic expression of lncARSR suppressed PTEN expression and induced Akt phosphorylation in A549 cells. The expression level of PTEN was higher in NSCLC samples than in adjacent non-tumor specimens. PTEN expression was negatively correlated with lncARSR in NSCLC specimens. Furthermore, we demonstrated that overexpression of lncARSR induced NSCLC cell growth and migration via regulating the PTEN/Akt signaling pathway. These results suggest that lncARSR acts as an oncogene in NSCLC development and could serve as a new potential therapeutic target.
Collapse
Affiliation(s)
- Jie Ying
- Clinical Research Center, Xuyi People’s HospitalXuyi 211700, Jiangsu, China
| | - Jian Yang
- Clinical Research Center, Xuyi People’s HospitalXuyi 211700, Jiangsu, China
| | - Yingzi Liu
- Department of Occupational Disease, Daqing People’s HospitalDaqing 163316, Heilongjiang, China
| |
Collapse
|
354
|
FAK Deficiency in Bone Marrow Stromal Cells Alters Their Homeostasis and Drives Abnormal Proliferation and Differentiation of Haematopoietic Stem Cells. Cells 2020; 9:cells9030646. [PMID: 32155953 PMCID: PMC7140540 DOI: 10.3390/cells9030646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/01/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022] Open
Abstract
Emerging evidence indicates that in myelodysplastic syndromes (MDS), the bone marrow (BM) microenvironment may also contribute to the ineffective, malignant haematopoiesis in addition to the intrinsic abnormalities of haematopoietic stem precursor cells (HSPCs). The BM microenvironment influences malignant haematopoiesis through indirect mechanisms, but the processes by which the BM microenvironment directly contributes to MDS initiation and progression have not yet been elucidated. Our previous data showed that BM-derived stromal cells (BMSCs) from MDS patients have an abnormal expression of focal adhesion kinase (FAK). In this study, we characterise the morpho-phenotypic features and the functional alterations of BMSCs from MDS patients and in FAK knock-downed HS-5 cells. The decreased expression of FAK or its phosphorylated form in BMSCs from low-risk (LR) MDS directly correlates with BMSCs' functional deficiency and is associated with a reduced level of haemoglobin. The downregulation of FAK in HS-5 cells alters their morphology, proliferation, and differentiation capabilities and impairs the expression of several adhesion molecules. In addition, we examine the CD34+ healthy donor (HD)-derived HSPCs' properties when co-cultured with FAK-deficient BMSCs. Both abnormal proliferation and the impaired erythroid differentiation capacity of HD-HSPCs were observed. Together, these results demonstrate that stromal adhesion mechanisms mediated by FAK are crucial for regulating HSPCs' homeostasis.
Collapse
|
355
|
NAFLD Preclinical Models: More than a Handful, Less of a Concern? Biomedicines 2020; 8:biomedicines8020028. [PMID: 32046285 PMCID: PMC7167756 DOI: 10.3390/biomedicines8020028] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of liver diseases ranging from simple steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and/or hepatocellular carcinoma. Due to its increasing prevalence, NAFLD is currently a major public health concern. Although a wide variety of preclinical models have contributed to better understanding the pathophysiology of NAFLD, it is not always obvious which model is best suitable for addressing a specific research question. This review provides insights into currently existing models, mainly focusing on murine models, which is of great importance to aid in the identification of novel therapeutic options for human NAFLD.
Collapse
|
356
|
Elucidating the mechanism of action of alpha-1-antitrypsin using retinal pigment epithelium cells exposed to high glucose. Potential use in diabetic retinopathy. PLoS One 2020; 15:e0228895. [PMID: 32032388 PMCID: PMC7006930 DOI: 10.1371/journal.pone.0228895] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alpha-1-antitrypsin is a protein involved in avoidance of different processes that are seen in diabetic retinopathy pathogenesis. These processes include apoptosis, extracellular matrix remodeling and damage of vessel walls and capillaries. Furthermore, because of its anti-inflammatory effects, alpha-1-antitrypsin has been proposed as a possible therapeutic approach for diabetic retinopathy. Our group tested alpha-1-antitrypsin in a type 1 diabetes mouse model and observed a reduction of inflammation and retinal neurodegeneration. Thus, shedding light on the mechanism of action of alpha-1-antitrypsin at molecular level may explain how it works in the diabetic retinopathy context and show its potential for use in other retinal diseases. METHODS In this work, we evaluated alpha-1-antitrypsin in an ARPE-19 human cell line exposed to high glucose. We explored the expression of different mediators on signaling pathways related to pro-inflammatory cytokines production, glucose metabolism, epithelial-mesenchymal transition and other proteins involved in the normal function of retinal pigment epithelium by RT-qPCR and Western Blot. RESULTS We obtained different expression patterns for evaluated mediators altered with high glucose exposure and corrected with the use of alpha-1-antitrypsin. CONCLUSIONS The expression profile obtained in vitro for the evaluated proteins and mRNA allowed us to explain our previous results obtained on mouse models and to hypothesize how alpha-1-antitrypsin hinder diabetic retinopathy progression on a complex network between different signaling pathways. GENERAL SIGNIFICANCE This network helps to understand the way alpha-1-antitrypsin works in diabetic retinopathy and its scope of action.
Collapse
|
357
|
Zhang S, Bai X, Shan F. The progress and confusion of anti-PD1/PD-L1 immunotherapy for patients with advanced non-small cell lung cancer. Int Immunopharmacol 2020; 80:106247. [PMID: 32007710 DOI: 10.1016/j.intimp.2020.106247] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/19/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Recently, immunotherapy has evolved into a true treatment modality with the approval of PD-1 and PD-L1 inhibitors as the standard care for first-line treatment in patients with non-small cell lung cancer (NSCLC). Until now, for patients with advanced NSCLC, treatment of targeting immune checkpoints reveals a promising survival benefit, and some patients even get long term survive, which creates a paradigm shift in NSCLC treatment. However, many issues or problems are also appearing in clinical practice, such as the lower overall efficacy rate (20-40%), treatment modes, populations choice of immunotherapy, drug resistance, and safety, etc. Thus, in this review, we will mainly summarize and discuss the recent development and confusion of PD-1/PD-L1 inhibitors for advanced NSCLC patients based on current clinical studies.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Chemoradiotherapy/methods
- Chemoradiotherapy/trends
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- ErbB Receptors/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Lung Neoplasms/immunology
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Molecular Targeted Therapy/methods
- Molecular Targeted Therapy/trends
- Mutation
- Neoplasm Staging
- Patient Selection
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Progression-Free Survival
- Randomized Controlled Trials as Topic
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/immunology
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Shuling Zhang
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang 110022, China; Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xueli Bai
- Department of Gynecology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110004, China; Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
358
|
Xing X, Guo S, Zhang G, Liu Y, Bi S, Wang X, Lu Q. miR-26a-5p protects against myocardial ischemia/reperfusion injury by regulating the PTEN/PI3K/AKT signaling pathway. ACTA ACUST UNITED AC 2020; 53:e9106. [PMID: 31994603 PMCID: PMC6984371 DOI: 10.1590/1414-431x20199106] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/10/2019] [Indexed: 01/08/2023]
Abstract
Reperfusion strategies in acute myocardial infarction (AMI) can cause a series of additional clinical damage, defined as myocardial ischemia/reperfusion (I/R) injury, and thus there is a need for effective therapeutic methods to attenuate I/R injury. miR-26a-5p has been proven to be an essential regulator for biological processes in different cell types. Nevertheless, the role of miR-26a-5p in myocardial I/R injury has not yet been reported. We established an I/R injury model in vitro and in vivo. In vitro, we used cardiomyocytes to simulate I/R injury using hypoxia/reoxygenation (H/R) assay. In vivo, we used C57BL/6 mice to construct I/R injury model. The infarct area was examined by TTC staining. The level of miR-26a-5p and PTEN was determined by bioinformatics methods, qRT-PCR, and western blot. In addition, the viability and apoptosis of cardiomyocytes were separately detected by MTT and flow cytometry. The targeting relationship between miR-26a-5p and PTEN was analyzed by the TargetScan website and luciferase reporter assay. I/R and H/R treatment induced myocardial tissue injury and cardiomyocyte apoptosis, respectively. The results showed that miR-26a-5p was down-regulated in myocardial I/R injury. PTEN was found to be a direct target of miR-26a-5p. Furthermore, miR-26a-5p effectively improved viability and inhibited apoptosis in cardiomyocytes upon I/R injury by inhibiting PTEN expression to activate the PI3K/AKT signaling pathway. miR-26a-5p could protect cardiomyocytes against I/R injury by regulating the PTEN/PI3K/AKT pathway, which offers a potential approach for myocardial I/R injury treatment.
Collapse
Affiliation(s)
- Xiaowei Xing
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Shuang Guo
- Department of Gastroenterology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Guanghao Zhang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yusheng Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Shaojie Bi
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Xin Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Qinghua Lu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
359
|
Tu T, Chen J, Chen L, Stiles BL. Dual-Specific Protein and Lipid Phosphatase PTEN and Its Biological Functions. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036301. [PMID: 31548229 DOI: 10.1101/cshperspect.a036301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) encodes a 403-amino acid protein with an amino-terminal domain that shares sequence homology with the actin-binding protein tensin and the putative tyrosine-protein phosphatase auxilin. Crystal structure analysis of PTEN has revealed a C2 domain that binds to phospholipids in membranes and a phosphatase domain that displays dual-specific activity toward both tyrosine (Y), serine (S)/threonine (T), as well as lipid substrates in vitro. Characterized primarily as a lipid phosphatase, PTEN plays important roles in multiple cellular processes including cell growth/survival as well as metabolism.
Collapse
Affiliation(s)
- Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Lulu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Bangyan L Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
360
|
Bhaskaran NA, Chevala NT, Kumar L. Nanopharmacokinetics, pharmacodynamics (PK/PD), and clinical relationship. NANOMEDICINES FOR BREAST CANCER THERANOSTICS 2020:245-268. [DOI: 10.1016/b978-0-12-820016-2.00011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
361
|
A review of predictive, prognostic and diagnostic biomarkers for brain tumours: towards personalised and targeted cancer therapy. JOURNAL OF RADIOTHERAPY IN PRACTICE 2019. [DOI: 10.1017/s1460396919000955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AbstractBackground:Brain tumours are relatively rare disease but present a large medical challenge as there is currently no method for early detection of the tumour and are typically not diagnosed until patients have progressed to symptomatic stage which significantly decreases chances of survival and also minimises treatment efficacy. However, if brain cancers can be diagnosed at early stages and also if clinicians have the potential to prospectively identify patients likely to respond to specific treatments, then there is a very high potential to increase patients’ treatment efficacy and survival. In recent years, there have been several investigations to identify biomarkers for brain cancer risk assessment, early detection and diagnosis, the likelihood of identifying which group of patients will benefit from a particular treatment and monitoring patient response to treatment.Materials and methods:This paper reports on a review of 21 current clinical and emerging biomarkers used in risk assessment, screening for early detection and diagnosis, and monitoring the response of treatment of brain cancers.Conclusion:Understanding biomarkers, molecular mechanisms and signalling pathways can potentially lead to personalised and targeted treatment via therapeutic targeting of specific genetic aberrant pathways which play key roles in malignant brain tumour formation. The future holds promising for the use of biomarker analysis as a major factor for personalised and targeted brain cancer treatment, since biomarkers have the potential to measure early disease detection and diagnosis, the risk of disease development and progression, improved patient stratification for various treatment paradigms, provide accurate information of patient response to a specific treatment and inform clinicians about the likely outcome of a brain cancer diagnosis independent of the treatment received.
Collapse
|
362
|
Beyond the Cell Surface: Targeting Intracellular Negative Regulators to Enhance T cell Anti-Tumor Activity. Int J Mol Sci 2019; 20:ijms20235821. [PMID: 31756921 PMCID: PMC6929154 DOI: 10.3390/ijms20235821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023] Open
Abstract
It is well established that extracellular proteins that negatively regulate T cell function, such as Cytotoxic T-Lymphocyte-Associated protein 4 (CTLA-4) and Programmed Cell Death protein 1 (PD-1), can be effectively targeted to enhance cancer immunotherapies and Chimeric Antigen Receptor T cells (CAR-T cells). Intracellular proteins that inhibit T cell receptor (TCR) signal transduction, though less well studied, are also potentially useful therapeutic targets to enhance T cell activity against tumor. Four major classes of enzymes that attenuate TCR signaling include E3 ubiquitin kinases such as the Casitas B-lineage lymphoma proteins (Cbl-b and c-Cbl), and Itchy (Itch), inhibitory tyrosine phosphatases, such as Src homology region 2 domain-containing phosphatases (SHP-1 and SHP-2), inhibitory protein kinases, such as C-terminal Src kinase (Csk), and inhibitory lipid kinases such as Src homology 2 (SH2) domain-containing inositol polyphosphate 5-phosphatase (SHIP) and Diacylglycerol kinases (DGKs). This review describes the mechanism of action of eighteen intracellular inhibitory regulatory proteins in T cells within these four classes, and assesses their potential value as clinical targets to enhance the anti-tumor activity of endogenous T cells and CAR-T cells.
Collapse
|
363
|
Chen Y, Chen D, Liu S, Yuan T, Guo J, Fang L, Du G. Systematic Elucidation of the Mechanism of Genistein against Pulmonary Hypertension via Network Pharmacology Approach. Int J Mol Sci 2019; 20:ijms20225569. [PMID: 31703458 PMCID: PMC6888439 DOI: 10.3390/ijms20225569] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Numerous studies have shown that genistein has a good therapeutic effect on pulmonary hypertension (PH). However, there has been no systematic research performed yet to elucidate its exact mechanism of action in relation to PH. In this study, a systemic pharmacology approach was employed to analyze the anti-PH effect of genistein. Firstly, the preliminary predicted targets of genistein against PH were obtained through database mining, and then the correlation of these targets with PH was analyzed. After that, the protein-protein interaction network was constructed, and the functional annotation and cluster analysis were performed to obtain the core targets and key pathways involved in exerting the anti-PH effect of genistein. Finally, the mechanism was further analyzed via molecular docking of genistein with peroxisome proliferator-activated receptor γ (PPARγ). The results showed that the anti-PH effect of genistein may be closely related to PPARγ, apoptotic signaling pathway, and the nitric oxide synthesis process. This study not only provides new insights into the mechanism of genistein against PH, but also provides novel ideas for network approaches for PH-related research.
Collapse
Affiliation(s)
- Yucai Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.C.); (S.L.)
| | - Di Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.); (T.Y.)
| | - Sijia Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.C.); (S.L.)
| | - Tianyi Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.); (T.Y.)
| | - Jian Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.C.); (S.L.)
- Correspondence: (J.G.); (L.F.); (G.D.)
| | - Lianhua Fang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.); (T.Y.)
- Correspondence: (J.G.); (L.F.); (G.D.)
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.); (T.Y.)
- Correspondence: (J.G.); (L.F.); (G.D.)
| |
Collapse
|
364
|
Chen T, Yu Q, Xin L, Guo L. Circular RNA circC3P1 restrains kidney cancer cell activity by regulating miR-21/PTEN axis and inactivating PI3K/AKT and NF- kB pathways. J Cell Physiol 2019; 235:4001-4010. [PMID: 31643094 DOI: 10.1002/jcp.29296] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
Kidney cancer (KC) seriously impacts public health. We detected the function and mechanism of circular RNA C3P1 (circC3P1) in KC cells. CCK-8, flow cytometry, migration, and invasion assay were respectively used to investigate the efficacies of circC3P1 and microRNA (miR)-21 on cell viability, apoptosis, migration, and invasion. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN), circC3P1, and miR-21 expression were changed by cell transfection and detected by quantitative reverse-transcription polymerase chain reaction. Moreover, the apoptosis/pathways-related proteins and proteins were detected by western blot analysis. Besides, the relation between PTEN and miR-21 was detected by luciferase assay. circC3P1 and PTEN were downregulated while miR-21 was upregulated in KC tissues. circC3P1 declined cell viability, migration, and invasion and caused apoptosis. Furthermore, circC3P1 negatively regulated miR-21; miR-21 mimic could reverse the efficacies of circC3P1. Besides, circC3P1 restrained the PI3K/AKT and NF-κB pathways by downregulating miR-21. Finally, PTEN was authenticated as a target of miR-21. circC3P1 restrained KC cell growth, migration, and invasion by regulating miR-21/PTEN axis and inactivating PI3K/AKT and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Tao Chen
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qinchao Yu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lei Xin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lei Guo
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
365
|
Rodríguez-Enríquez S, Marín-Hernández Á, Gallardo-Pérez JC, Pacheco-Velázquez SC, Belmont-Díaz JA, Robledo-Cadena DX, Vargas-Navarro JL, Corona de la Peña NA, Saavedra E, Moreno-Sánchez R. Transcriptional Regulation of Energy Metabolism in Cancer Cells. Cells 2019; 8:cells8101225. [PMID: 31600993 PMCID: PMC6830338 DOI: 10.3390/cells8101225] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 01/17/2023] Open
Abstract
Cancer development, growth, and metastasis are highly regulated by several transcription regulators (TRs), namely transcription factors, oncogenes, tumor-suppressor genes, and protein kinases. Although TR roles in these events have been well characterized, their functions in regulating other important cancer cell processes, such as metabolism, have not been systematically examined. In this review, we describe, analyze, and strive to reconstruct the regulatory networks of several TRs acting in the energy metabolism pathways, glycolysis (and its main branching reactions), and oxidative phosphorylation of nonmetastatic and metastatic cancer cells. Moreover, we propose which possible gene targets might allow these TRs to facilitate the modulation of each energy metabolism pathway, depending on the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Norma Angélica Corona de la Peña
- Unidad de Investigación Médica en Trombosis, Hemostasia y Aterogénesis, Hospital General Regional Carlos McGregor-Sánchez, México CP 03100, Mexico.
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| | - Rafael Moreno-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| |
Collapse
|
366
|
Holly JMP, Biernacka K, Perks CM. The Neglected Insulin: IGF-II, a Metabolic Regulator with Implications for Diabetes, Obesity, and Cancer. Cells 2019; 8:cells8101207. [PMID: 31590432 PMCID: PMC6829378 DOI: 10.3390/cells8101207] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
When originally discovered, one of the initial observations was that, when all of the insulin peptide was depleted from serum, the vast majority of the insulin activity remained and this was due to a single additional peptide, IGF-II. The IGF-II gene is adjacent to the insulin gene, which is a result of gene duplication, but has evolved to be considerably more complicated. It was one of the first genes recognised to be imprinted and expressed in a parent-of-origin specific manner. The gene codes for IGF-II mRNA, but, in addition, also codes for antisense RNA, long non-coding RNA, and several micro RNA. Recent evidence suggests that each of these have important independent roles in metabolic regulation. It has also become clear that an alternatively spliced form of the insulin receptor may be the principle IGF-II receptor. These recent discoveries have important implications for metabolic disorders and also for cancer, for which there is renewed acknowledgement of the importance of metabolic reprogramming.
Collapse
Affiliation(s)
- Jeff M P Holly
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Kalina Biernacka
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Claire M Perks
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
367
|
Okumura S, Hirano Y, Komatsu Y. Inhibition of breast cancer cell proliferation with anti-microRNA oligonucleotides flanked by interstrand cross-linked duplexes. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 39:225-235. [PMID: 31583946 DOI: 10.1080/15257770.2019.1671595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most frequent cancer affecting women worldwide. Traditional chemotherapy, hormone therapy, and targeted therapy are used for breast cancer treatment. However, breast cancer is a heterogeneous disease, and patients often develop drug resistance. Therefore, various new therapeutic strategies have been investigated, including microRNA regulation. Anti-microRNA oligonucleotides (AMOs) are one of the most potent agents in oligonucleotide therapy. The inhibition activity of an AMO can be increased by flanking its single-stranded antisense sequence (the widely used structure for AMOs) with interstrand cross-linked duplexes (CLDs). An extrastable CLD improves nuclease resistance and stabilizes hybridization with a target. This study investigated the effects of anti-microRNA-21 (miR-21) AMO modified with CLDs on breast cancer cells without using reporter assay. The CLD-modified AMO suppressed breast cancer cell proliferation for a long duration compared to other types of AMOs. In addition, it expectedly up-regulated the miR-21-controlled expression of tumor suppressor genes. Therefore, an AMO flanked by CLDs can be a promising strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Sho Okumura
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Cosmo Bio Co., Ltd, Otaru, Japan.,Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, Japan
| | - Yu Hirano
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, Japan
| | - Yasuo Komatsu
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
368
|
Frazier TW. Autism Spectrum Disorder Associated with Germline Heterozygous PTEN Mutations. Cold Spring Harb Perspect Med 2019; 9:a037002. [PMID: 31307976 PMCID: PMC6771360 DOI: 10.1101/cshperspect.a037002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This review examines our current understanding of autism spectrum disorder (ASD), its prevalence, impact, behavioral treatment, and outcomes. Building on this knowledge, ASD associated with PTEN mutations is introduced and recent human studies of neurobehavioral and neuroimaging findings in patients with PTEN mutations with and without ASD are reviewed. In doing so, we present evidence supporting a model of PTEN loss leading to neurobehavioral deficits, including ASD and intellectual disability. Next, we describe the neurobehavioral spectrum observed across PTEN mutation cases, adding specificity where possible, based on data from recent studies of child and adult PTEN patients. Finally, we end with a discussion of potential clinical recommendations for improving interventions and supports for people with PTEN-ASD and future research avenues for understanding and treating the functional and cognitive deficits in PTEN-ASD.
Collapse
|
369
|
Gerisch G, Prassler J, Butterfield N, Ecke M. Actin Waves and Dynamic Patterning of the Plasma Membrane. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:397-411. [PMID: 31543704 PMCID: PMC6747932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Plasma membrane and underlying actin network are connected to a functional unit that by non-linear interactions is capable of forming patterns. For instance, in cell motility and chemotaxis, cells polarize to form a protruding front and a retracting tail. Here we address dynamic patterns that are formed on a planar substrate surface and are therefore easily accessible to optical recording. In these patterns two distinct areas of the membrane and actin cortex are interconverted at the site of circular actin waves. The inner territory circumscribed by a wave is distinguished from the external area by a high PIP3 content and high Ras activity. In contrast, the external area is occupied with the PIP3-degrading phosphatase PTEN. In the underlying cortex, these areas differ in the proteins associated with the actin network. Actin waves can be formed at zones of increasing as well as decreasing Ras activity. Both types of waves are headed by myosin IB. When waves collide, they usually extinguish each other, and their decay is accompanied by the accumulation of coronin. No membrane patterns have been observed after efficient depolymerization of actin, suggesting that residual actin filaments are necessary for the pattern generating system to work. Where appropriate, we relate the experimental data obtained with Dictyostelium to human normal and malignant cell behavior, in particular to the role of Ras-GAP as an enhancer of macropinocytosis, to mutations in the tumor suppressor PTEN, to frustrated phagocytosis, and to the role of coronin in immune cells and neurons.
Collapse
Affiliation(s)
- Guenther Gerisch
- To whom all correspondence should be addressed: Dr. Günther Gerisch, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany; Tel: +49 89 8578-2326, Fax: +49 89 8578-3885,
| | | | | | | |
Collapse
|
370
|
Xiang X, Zhuang L, Chen H, Yang X, Li H, Li G, Yu J. Everolimus inhibits the proliferation and migration of epidermal growth factor receptor-resistant lung cancer cells A549 via regulating the microRNA-4328/phosphatase and tensin homolog signaling pathway. Oncol Lett 2019; 18:5269-5276. [PMID: 31612036 PMCID: PMC6781784 DOI: 10.3892/ol.2019.10887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/19/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the most common cancer type worldwide, and investigating novel therapeutics methods for the treatment of chemoresistant lung cancer are of notable clinical significance. Reverse transcription-quantitative polymerase chain reaction and western blotting assays were performed to analyze the expression levels of phosphatase and tensin homolog (PTEN) and microRNA-4328 (miR-4328), and Cell Counting Kit-8 (CCK-8) and Transwell migration assays were conducted to evaluate the proliferation and migration of A549 cells, respectively. Everolimus was observed to upregulate the expression of PTEN and inhibit the proliferation and migration of A549 cells in a dose-dependent manner. The knockdown of PTEN abolished the effects of everolimus on the proliferation and migration of A549 cells, and everolimus was demonstrated to upregulate PTEN, and inhibit the proliferation and migration of A549 cells via downregulating miR-4328. Collectively, the results of the present study indicate that everolimus inhibited the proliferation and migration of EGFR-resistant A549 lung cancer cells via regulating the miR-4328/PTEN signaling pathway.
Collapse
Affiliation(s)
- Xudong Xiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Li Zhuang
- Department of Palliative Medicine and Palliative Medicine Research Center, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Huicheng Chen
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Xiumei Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Heng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Gaofeng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan 650118, P.R. China
| | - Jing Yu
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
371
|
Hjelholt AJ, Lee KY, Arlien-Søborg MC, Pedersen SB, Kopchick JJ, Puri V, Jessen N, Jørgensen JOL. Temporal patterns of lipolytic regulators in adipose tissue after acute growth hormone exposure in human subjects: A randomized controlled crossover trial. Mol Metab 2019; 29:65-75. [PMID: 31668393 PMCID: PMC6731350 DOI: 10.1016/j.molmet.2019.08.013] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/11/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022] Open
Abstract
Objective Growth hormone (GH) stimulates lipolysis, but the underlying mechanisms remain incompletely understood. We examined the effect of GH on the expression of lipolytic regulators in adipose tissue (AT). Methods In a randomized, placebo-controlled, cross-over study, nine men were examined after injection of 1) a GH bolus and 2) a GH-receptor antagonist (pegvisomant) followed by four AT biopsies. In a second study, eight men were examined in a 2 × 2 factorial design including GH infusion and 36-h fasting with AT biopsies obtained during a basal period and a hyperinsulinemic-euglycemic clamp. Expression of GH-signaling intermediates and lipolytic regulators were studied by PCR and western blotting. In addition, mechanistic experiments in mouse models and 3T3-L1 adipocytes were performed. Results The GH bolus increased circulating free fatty acids (p < 0.0001) together with phosphorylation of signal transducer and activator of transcription 5 (STAT5) (p < 0.0001) and mRNA expression of the STAT5-dependent genes cytokine-inducible SH2-containing protein (CISH) and IGF-1 in AT. This was accompanied by suppressed mRNA expression of G0/G1 switch gene 2 (G0S2) (p = 0.007) and fat specific protein 27 (FSP27) (p = 0.002) and upregulation of phosphatase and tensin homolog (PTEN) mRNA expression (p = 0.03). Suppression of G0S2 was also observed in humans after GH infusion and fasting, as well as in GH transgene mice, and in vitro studies suggested MEK-PPARγ signaling to be involved. Conclusions GH-induced lipolysis in human subjects in vivo is linked to downregulation of G0S2 and FSP27 and upregulation of PTEN in AT. Mechanistically, in vitro data suggest that GH acts via MEK to suppress PPARγ-dependent transcription of G0S2. ClinicalTrials.govNCT02782221 and NCT01209429. Acute GH exposure in human subjects in vivo stimulates lipolysis and release of FFA together with GH signaling in adipose tissue. GH-induced lipolysis is associated with suppression of G0S2 and FSP27 and upregulation of PTEN in human subjects in vivo. Inhibition of MEK and activation of PPARγ abrogate GH-induced suppression of G0S2 mRNA expression in vitro.
Collapse
Affiliation(s)
- Astrid Johannesson Hjelholt
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark.
| | - Kevin Y Lee
- Heritage College of Osteopathic Medicine, Ohio University, 204 Grosvenor Hall, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Konneker Research Center 108, Athens, OH 45701, USA
| | - Mai Christiansen Arlien-Søborg
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark
| | - Steen Bønløkke Pedersen
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 2., 8200 Aarhus N, Denmark
| | - John J Kopchick
- Heritage College of Osteopathic Medicine, Ohio University, 204 Grosvenor Hall, Athens, OH 45701, USA; The Edison Biotechnology Institute, Ohio University, Konneker Research Center, 172 Water Tower Dr., Athens, OH 45701, USA
| | - Vishwajeet Puri
- Heritage College of Osteopathic Medicine, Ohio University, 204 Grosvenor Hall, Athens, OH 45701, USA; The Diabetes Institute, Ohio University, Konneker Research Center 108, Athens, OH 45701, USA
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Hedeager 3, 2., 8200 Aarhus N, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Wilh. Meyers Allé 4, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, Vennelyst Boulevard 4, 8000 Aarhus C, Denmark
| | - Jens Otto L Jørgensen
- Medical Research Laboratory, Department of Clinical Medicine, Endocrinology and Internal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200 Aarhus N, Denmark
| |
Collapse
|
372
|
Zhuang Z, Yu D, Chen Z, Liu D, Yuan G, Yirong N, Sun L, Liu Y, He R, Wang K. Curcumin Inhibits Joint Contracture through PTEN Demethylation and Targeting PI3K/Akt/mTOR Pathway in Myofibroblasts from Human Joint Capsule. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:4301238. [PMID: 31511778 PMCID: PMC6712967 DOI: 10.1155/2019/4301238] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/27/2019] [Accepted: 07/14/2019] [Indexed: 12/25/2022]
Abstract
Joint contracture is increasingly regarded as a clinical problem that leads to irreversible dysfunction of the joint. It is a pathophysiological process following joint injury, which is marked by the activation of myofibroblasts. There is currently no effective treatment for the prevention of joint contracture. Curcumin is a polyphenol pigment extracted from turmeric, which possesses anti-inflammatory, antioxidative, and antitumor properties. In the present study, we demonstrated that curcumin exerts a protective effect against joint contracture via the inhibition of myofibroblast proliferation and migration in a time- and concentration-dependent manner. Moreover, we indicated that phosphatase and tension homolog (PTEN) was downregulated in myofibroblasts in vitro and in the contracture capsule tissues of patients in vivo. Additionally, western blot analysis revealed a negative correlation between the expression levels of PTEN and the fibrosis marker protein alpha smooth muscle cell actin. Methylation-specific PCR results suggested that curcumin was able to demethylate PTEN in a similar manner to the demethylation agent 5-azacytidine, increasing PTEN expression and further inhibiting phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling. In conclusion, our data illustrate part of the mechanism of curcumin inhibition in joint contracture. These results support the hypothesis that curcumin may potentially be used as a novel candidate for the treatment of joint contracture.
Collapse
Affiliation(s)
- Ze Zhuang
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Dongjie Yu
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Zheng Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China
| | - Dezhao Liu
- Departments of Anesthesiolgy, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Guohui Yuan
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Ni Yirong
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou 510631, Guangdong, China
| | - Linlin Sun
- Departments of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Yuangao Liu
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Ronghan He
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Kun Wang
- Departments of Joint Surgery and Orthopedic Trauma, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
373
|
Tan S, Khumalo N, Bayat A. Understanding Keloid Pathobiology From a Quasi-Neoplastic Perspective: Less of a Scar and More of a Chronic Inflammatory Disease With Cancer-Like Tendencies. Front Immunol 2019; 10:1810. [PMID: 31440236 PMCID: PMC6692789 DOI: 10.3389/fimmu.2019.01810] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 07/17/2019] [Indexed: 01/01/2023] Open
Abstract
Keloids are considered as benign fibroproliferative skin tumors growing beyond the site of the original dermal injury. Although traditionally viewed as a form of skin scarring, keloids display many cancer-like characteristics such as progressive uncontrolled growth, lack of spontaneous regression and extremely high rates of recurrence. Phenotypically, keloids are consistent with non-malignant dermal tumors that are due to the excessive overproduction of collagen which never metastasize. Within the remit of keloid pathobiology, there is increasing evidence for the various interplay of neoplastic-promoting and suppressing factors, which may explain its aggressive clinical behavior. Amongst the most compelling parallels between keloids and cancer are their shared cellular bioenergetics, epigenetic methylation profiles and epithelial-to-mesenchymal transition amongst other disease biological (genotypic and phenotypic) behaviors. This review explores the quasi-neoplastic or cancer-like properties of keloids and highlights areas for future study.
Collapse
Affiliation(s)
- Silvian Tan
- Plastic and Reconstructive Surgery Research, Centre for Dermatology Research, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom
| | - Nonhlanhla Khumalo
- Hair and Skin Research Laboratory, Department of Dermatology, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- Plastic and Reconstructive Surgery Research, Centre for Dermatology Research, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom
- Hair and Skin Research Laboratory, Department of Dermatology, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
374
|
LHPP suppresses bladder cancer cell proliferation and growth via inactivating AKT/p65 signaling pathway. Biosci Rep 2019; 39:BSR20182270. [PMID: 31262971 PMCID: PMC6667728 DOI: 10.1042/bsr20182270] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
Bladder cancer (BC) is one of the commonest malignancies in the urinary system. Recent evidences have shown that Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) serves as a tumor suppressor in hepatocellular carcinoma and cervical cancer. However, little is known about its function in BC. Here, we aimed to investigate the role of LHPP in BC. We found that LHPP was down-regulated in BC tissues and cells. Knockdown of LHPP promoted the proliferation and growth of BC cells T24 and 5637. Inverse results were observed in SW780 and BIU87 cells with ectopic LHPP expression. LHPP also repressed the glycolysis of BC cells. At the molecular level, LHPP silencing led to enhanced phosphorylation of both AKT and p65, as well as up-regulation of their downstream targets Bcl-2 and Cyclin D1. Inhibition of AKT by MK2206 blunted the increased phosphorylation of p65 caused by LHPP knockdown, suggesting that LHPP silencing activated p65 through AKT. Importantly, p65 inhibitor (caffeic acid phenethyl ester) exhibited larger suppressive effect on the proliferation of LHPP knockdown BC cells as compared with Ctrl cell. Our study demonstrates that LHPP suppresses BC cell growth via inactivating AKT/p65 signaling pathway.
Collapse
|
375
|
Yu Y, Xiong Y, Ladeiras D, Yang Z, Ming XF. Myosin 1b Regulates Nuclear AKT Activation by Preventing Localization of PTEN in the Nucleus. iScience 2019; 19:39-53. [PMID: 31349190 PMCID: PMC6660601 DOI: 10.1016/j.isci.2019.07.010] [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: 12/11/2018] [Revised: 05/17/2019] [Accepted: 07/05/2019] [Indexed: 02/08/2023] Open
Abstract
Insulin-induced AKT activation is dependent on phosphoinositide 3-kinase and opposed by tumor suppressor phosphatase and tensin homolog (PTEN). Our previous study demonstrates that myosin 1b (MYO1B) mediates arginase-II-induced activation of mechanistic target of rapamycin complex 1 that is regulated by AKT. However, the role of MYO1B in AKT activation is unknown. Here we show that silencing MYO1B in mouse embryonic fibroblasts (MEF) inhibits insulin-induced nuclear but not cytoplasmic AKT activation accompanied by elevated nuclear PTEN level. Co-immunoprecipitation, co-immunostaining, and proximity ligation assay show an interaction of MYO1B and PTEN resulting in reduced nuclear PTEN. Moreover, the elevated nuclear PTEN upon silencing MYO1B promotes apoptosis of MEFs and melanoma B16F10 cells. Taken together, we demonstrate that MYO1B, by interacting with PTEN, prevents nuclear localization of PTEN contributing to nuclear AKT activation and suppression of cell apoptosis. This may present a therapeutic approach for cancer treatment such as melanoma. MYO1B, by interacting with PTEN, prevents PTEN localization in the nucleus MYO1B prevents nuclear localization of PTEN depending on its motor activity This contributes to nuclear AKT activation and suppression of cell apoptosis Targeting MYO1B may represent a therapeutic approach for cancer treatment
Collapse
Affiliation(s)
- Yi Yu
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Yuyan Xiong
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Diogo Ladeiras
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Zhihong Yang
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland.
| | - Xiu-Fen Ming
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland.
| |
Collapse
|
376
|
Burgy M, Chenard MP, Noël G, Bourahla K, Schott R. Bone metastases from a 1p/19q codeleted and IDH1-mutant anaplastic oligodendroglioma: a case report. J Med Case Rep 2019; 13:202. [PMID: 31248444 PMCID: PMC6598291 DOI: 10.1186/s13256-019-2061-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 03/20/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Oligodendroglioma is a rare type of primary brain tumor which, like other malignant gliomas, metastasizes very rarely even when in high-grade form. CASE REPORT A 36-year-old white man diagnosed 29 months previously as having 1p/19q codeleted anaplastic oligodendroglioma presented bilateral cruralgia and lower limb motor deficits. A computed tomography scan showed multiple osteoblastic bone lesions. The presence of oligodendroglial cells was revealed by bone marrow biopsy and confirmed by immunohistochemical analyses. A positon emission tomography-computed tomography scan confirmed the exclusive involvement of bones. CONCLUSION This case joins less than 20 other reported cases of oligodendroglioma bone marrow metastasis, and is one of only a handful of cases of diffuse bone metastases beyond the axial skeleton. To the best of our knowledge, the early relapse of 1p/19q codeleted anaplastic oligodendroglioma with this distribution of metastases has never been described in the literature.
Collapse
Affiliation(s)
- Mickaël Burgy
- Medical Oncology Department, Centre Paul Strauss, 3 Rue de la Porte de l'Hôpital, 67000, Strasbourg, France.,Université de Strasbourg, LBP, CNRS UMR 7213, Illkirch, France
| | | | - Georges Noël
- Radiotherapy Department, Centre Paul-Strauss, Strasbourg, France
| | - Khalil Bourahla
- Nuclear Medicine Department, Centre Paul-Strauss, Strasbourg, France
| | - Roland Schott
- Medical Oncology Department, Centre Paul Strauss, 3 Rue de la Porte de l'Hôpital, 67000, Strasbourg, France.
| |
Collapse
|
377
|
Pilarski R. PTEN Hamartoma Tumor Syndrome: A Clinical Overview. Cancers (Basel) 2019; 11:cancers11060844. [PMID: 31216739 PMCID: PMC6627214 DOI: 10.3390/cancers11060844] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/12/2022] Open
Abstract
The phosphatase and tensin homolog (PTEN) hamartoma tumor syndrome (PHTS) is a grouping of related genetic disorders that has been linked to germline mutations in the PTEN gene. These disorders include Cowden syndrome (CS), Bannayan–Riley–Ruvalcaba syndrome, adult Lhermitte–Duclos disease, and autism spectrum disorders associated with macrocephaly. The majority of the clinical information available on PHTS, however, is related to individuals diagnosed with CS. There is still much to be learned about this disorder, since diagnostic criteria for CS were only established in 1996, before the identification of the PTEN gene, and were based primarily on features seen in cases reported in the existing literature. More recently, however, data from several large series of patients have shown that a number of the clinical features associated with PTEN mutations are either more or less common than previously reported. In addition, we now know that only about 30–35% of patients meeting clinical diagnostic criteria for Cowden syndrome actually have a detectable PTEN mutation. Thus, our understanding of PTEN-related diseases and their management has evolved significantly over time. The United States National Comprehensive Cancer Network (NCCN) has produced and regularly updates practice guidelines which include clinical diagnostic criteria as well as guidelines for PTEN testing and management of patients with mutations. This review will summarize the overall literature on PHTS as well as recent findings which are broadening our understanding of this set of disorders.
Collapse
Affiliation(s)
- Robert Pilarski
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43221, USA.
| |
Collapse
|
378
|
Mei M, Zhang M. Non-coding RNAs in Natural Killer/T-Cell Lymphoma. Front Oncol 2019; 9:515. [PMID: 31263681 PMCID: PMC6584837 DOI: 10.3389/fonc.2019.00515] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022] Open
Abstract
Natural killer/T-cell lymphoma (NKTCL) is a rare and aggressive subtype of non-Hodgkin's lymphoma that is associated with a poor outcome. Non-coding RNAs (ncRNAs), which account for 98% of human RNAs, lack the function of encoding proteins but instead have the important function of regulating gene expression, including transcription, translation, RNA splicing, editing, and turnover. However, the roles and mechanisms of aberrantly expressed ncRNAs in NKTCL are not fully clear. Aberrant expressions of microRNA (miRNAs) affect the PI3K/AKT signaling pathways (miRNA-21, miRNA-155, miRNA-150, miRNA-142, miRNA-494), NF-κB (miRNA-146a, miRNA-155) and cell cycle signaling pathways to regulate cell function. Moreover, Epstein-Barr virus (EBV) encoded miRNAs and EBV oncoprotein LMP-1 regulated the expression of cellular genes that induce invasion, metastasis, cell cycle progression and cellular transformation. In addition, NKTCL-associated Long non-coding RNA (lncRNA) ZFAS1 regulated certain pathways and lncRNA MALAT1 acted as a predictive marker. This review article provides an overview of ncRNAs associated with NKTCL, summarizes the function of significantly differentially expressed hotspot non-coding RNAs that contribute to the pathogenesis, diagnoses, treatment and prognosis of NKTCL and discusses the relevance of these ncRNAs to clinical practice.
Collapse
Affiliation(s)
- Mei Mei
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
379
|
Zheng H, Liu J, Tycksen E, Nunley R, McAlinden A. MicroRNA-181a/b-1 over-expression enhances osteogenesis by modulating PTEN/PI3K/AKT signaling and mitochondrial metabolism. Bone 2019; 123:92-102. [PMID: 30898695 PMCID: PMC6491221 DOI: 10.1016/j.bone.2019.03.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/14/2022]
Abstract
MicroRNAs are small non-coding RNAs that play important roles in many cellular processes including proliferation, metabolism and differentiation. They function by binding to specific regions within the 3'UTR of target mRNAs resulting in suppression of protein synthesis and modulation of potentially many cellular pathways. We previously showed that miRNA expression levels differed between cells from distinct regions of developing human embryonic long bones. Specifically, we found that miR-181a-1 was significantly more highly expressed in hypertrophic chondrocytes compared to proliferating differentiated or progenitor chondrocytes, suggesting a potential role in regulating chondrocyte hypertrophy and/or endochondral bone formation. The goal of this study was to determine how miR-181a-1 together with its clustered miRNA, miR-181b-1, regulates osteogenesis. We show that over-expression of the miR-181a/b-1 cluster enhanced osteogenesis and that cellular pathways associated with protein synthesis and mitochondrial metabolism were significantly up-regulated. Metabolic assays revealed that the oxygen consumption rate and ATP-linked respiration were increased by miR-181a/b-1. To further decipher a potential mechanism causing these metabolic changes, we showed that PTEN (phosphatase and tensin homolog) levels were suppressed following miR-181a/b-1 over-expression, and that PI3K/AKT signaling was subsequently increased. Over-expression of PTEN was found to attenuate the enhancing effects of miR-181a/b-1, providing further evidence that miR-181a/b-1 regulates the PTEN/PI3K/AKT axis to enhance osteogenic differentiation and mitochondrial metabolism. These findings have important implications for the design of miR-181a/b targeting strategies to treat bone conditions such as fractures or heterotopic ossification.
Collapse
Affiliation(s)
- Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Jin Liu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Eric Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St Louis, MO, United States of America.
| | - Ryan Nunley
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, United States of America; Shriners Hospital for Children - St Louis, St Louis, MO, United States of America.
| |
Collapse
|
380
|
Wu SM, Li TH, Yun H, Ai HW, Zhang KH. miR-140-3p Knockdown Suppresses Cell Proliferation and Fibrogenesis in Hepatic Stellate Cells via PTEN-Mediated AKT/mTOR Signaling. Yonsei Med J 2019; 60:561-569. [PMID: 31124340 PMCID: PMC6536388 DOI: 10.3349/ymj.2019.60.6.561] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/07/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Liver fibrosis is a major cause of morbidity and mortality and the outcome of various chronic liver diseases. Activation of hepatic stellate cells (HSCs) is the key event in liver fibrosis. Studies have confirmed that miR-140-3p plays a potential regulatory effect on HSC activation. However, whether miR-140-3p mediates the liver fibrosis remains unknown. MATERIALS AND METHODS Expression of miR-140-3p was detected by real-time quantitative PCR (qPCR). Cell proliferation was measured by MTT, while cell apoptosis rate was determined via flow cytometry. Western blot assay was used to detect the expression of cleaved PARP. The fibrogenic effect was evaluated by expression of α-smooth muscle actin and desmin. Functional experiments were performed in transforming growth factor β1 (TGF-β1)-induced HSC-T6 cells with transfection of anti-miR-140-3p and/or siPTEN. Target binding between miR-140-3p and PTEN was predicted by the TargetScan database and identified using luciferase reporter assay and RNA immunoprecipitation. RESULTS TGF-β1 induced the activation of HSC-T6 cells, and miR-140-3p expression varied according to HSC-T6 cell activation status. Knockdown of miR-140-3p reduced cell proliferation and the expressions of α-SMA and desmin, as well as increased apoptosis, in TGF-β1-induced HSC-T6 cells, which could be blocked by PTEN silencing. Additionally, inactivation of the AKT/mTOR signaling pathway stimulated by miR-140-3p knockdown was abolished when silencing PTEN expression. PTEN was negatively regulated by miR-140-3p via direct binding in HSC-T6 cells. CONCLUSION miR-140-3p is an important mediator in HSC-T6 cell activation, and miR-140-3p knockdown suppresses cell proliferation and fibrogenesis in TGF-β1-induced HSC-T6 cells, indicating that miR-140-3p may be a potential novel molecular target for liver fibrosis.
Collapse
Affiliation(s)
- Shi Min Wu
- Wuhan Center for Clinical Laboratory, Wuhan Forth Hospital; Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Hong Li
- Department of Ophthalmology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Yun
- Wuhan Center for Clinical Laboratory, Wuhan Forth Hospital; Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Wu Ai
- Department of Clinical Laboratory, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Hui Zhang
- Wuhan Center for Clinical Laboratory, Wuhan Forth Hospital; Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
381
|
Su M, Xiao Y, Ma J, Tang Y, Tian B, Zhang Y, Li X, Wu Z, Yang D, Zhou Y, Wang H, Liao Q, Wang W. Circular RNAs in Cancer: emerging functions in hallmarks, stemness, resistance and roles as potential biomarkers. Mol Cancer 2019; 18:90. [PMID: 30999909 PMCID: PMC6471953 DOI: 10.1186/s12943-019-1002-6] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of RNA molecules with closed loops and high stability. CircRNAs are abundantly expressed in eukaryotic organisms and exhibit both location- and step-specificity. In recent years, circRNAs are attracting considerable research attention attributed to their possible contributions to gene regulation through a variety of actions, including sponging microRNAs, interacting with RNA-binding proteins, regulating transcription and splicing, and protein translation. Growing evidence has revealed that circRNAs play critical roles in the development and progression of diseases, especially in cancers. Without doubt, expanding our understanding of circRNAs will enrich knowledge of cancer and provide new opportunities for cancer therapy. In this review, we provide an overview of the characteristics, functions and functional mechanisms of circRNAs. In particular, we summarize current knowledge regarding the functions of circRNAs in the hallmarks, stemness, resistance of cancer, as well as the possibility of circRNAs as biomarkers in cancer.
Collapse
Affiliation(s)
- Min Su
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of the Central Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yuhang Xiao
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410001, People's Republic of China
| | - Junliang Ma
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yanyan Tang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Tian
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yuqin Zhang
- Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410001, People's Republic of China
| | - Xu Li
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhining Wu
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Desong Yang
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yong Zhou
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,Department of the Central Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| | - Wenxiang Wang
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
382
|
Qin Y, Hu Q, Xu J, Ji S, Dai W, Liu W, Xu W, Sun Q, Zhang Z, Ni Q, Zhang B, Yu X, Xu X. PRMT5 enhances tumorigenicity and glycolysis in pancreatic cancer via the FBW7/cMyc axis. Cell Commun Signal 2019; 17:30. [PMID: 30922330 PMCID: PMC6440122 DOI: 10.1186/s12964-019-0344-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The epigenetic factor protein arginine methyltransferase 5 (PRMT5) has been reported to play vital roles in a wide range of cellular processes, such as gene transcription, genomic organization, differentiation and cell cycle control. However, its role in pancreatic cancer remains unclear. Our study aimed to investigate the roles of PRMT5 in pancreatic cancer prognosis and progression and to explore the underlying molecular mechanism. METHODS Real-time PCR, immunohistochemistry and analysis of a dataset from The Cancer Genome Atlas (TCGA) were performed to study the expression of PRMT5 at the mRNA and protein levels in pancreatic cancer. Cell proliferation assays, including cell viability, colony formation ability and subcutaneous mouse model assays, were utilized to confirm the role of PRMT5 in cell proliferation and tumorigenesis. A Seahorse extracellular flux analyzer, a glucose uptake kit, a lactate level measurement kit and the measurement of 18F-FDG (fluorodeoxyglucose) uptake by PET/CT (positron emission tomography/computed tomography) imaging were used to verify the role of PRMT5 in aerobic glycolysis, which sustains cell proliferation. The regulatory effect of PRMT5 on cMyc, a master regulator of oncogenesis and aerobic glycolysis, was explored by quantitative PCR and protein stability measurements. RESULTS PRMT5 expression was significantly upregulated in pancreatic cancer tissues compared with that in adjacent normal tissues. Clinically, elevated expression of PRMT5 was positively correlated with worse overall survival in pancreatic cancer patients. Silencing PRMT5 expression inhibited the proliferation of pancreatic cancer cells both in vitro and in vivo. Moreover, PRMT5 regulated aerobic glycolysis in vitro in cell lines, in vivo in pancreatic cancer patients and in a xenograft mouse model used to measure 18F-FDG uptake. We found that mechanistically, PRMT5 posttranslationally regulated cMyc stability via F-box/WD repeat-containing protein 7 (FBW7), an E3 ubiquitin ligase that controls cMyc degradation. Moreover, PRMT5 epigenetically regulated the expression of FBW7 in pancreatic cancer cells. CONCLUSIONS The present study demonstrated that PRMT5 epigenetically silenced the expression of the tumor suppressor FBW7, leading to increased cMyc levels and the subsequent enhancement of the proliferation of and aerobic glycolysis in pancreatic cancer cells. The PRMT5/FBW7/cMyc axis could be a potential therapeutic target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Weixing Dai
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Qiqing Sun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China
| |
Collapse
|
383
|
Abstract
AbstractDairy cows with fatty liver or ketosis display decreased insulin sensitivity and defects in the insulin receptor substrate (IRS)/PI3K/AKT signaling pathway. Phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor and also a negative regulator of insulin signaling and peripheral insulin sensitivity. We investigated the hypothesis that PTEN may affect the insulin pathway-mediated hepatic glucose and lipid metabolism in dairy cows. Adenovirus vectors that over-express and silence PTEN were constructed, and then transfected into hepatocytes isolated from calves to investigate the effect of PTEN on PI3K/AKT signaling pathway. PTEN silencing increased the phosphorylation of AKT and the expression of PI3K but decreased the phosphorylation of IRS1, which increased the phosphorylation levels of glycogen synthase kinase-3β (GSK-3β) and expression of sterol regulatory element-binding protein-1c (SREBP-1c). Increased GSK-3β phosphorylation further up-regulated expression of the key enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6-Pase) involved in gluconeogenesis. Furthermore, the expression of SREBP-1c target gene fatty acid synthase (FAS) also increased significantly. We further showed that PTEN over-expression could reverse the above results. PTEN negatively regulates the enzymes involved in hepatic gluconeogenesis and lipid synthesis, which suggests that PTEN may be a therapeutic target for ketosis and fatty liver in dairy cows.
Collapse
|
384
|
Protein Phosphatases-A Touchy Enemy in the Battle Against Glioblastomas: A Review. Cancers (Basel) 2019; 11:cancers11020241. [PMID: 30791455 PMCID: PMC6406705 DOI: 10.3390/cancers11020241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor arising from brain parenchyma. Although many efforts have been made to develop therapies for GBM, the prognosis still remains poor, mainly because of the difficulty in total resection of the tumor mass from brain tissue and the resistance of the residual tumor against standard chemoradiotherapy. Therefore, novel adjuvant therapies are urgently needed. Recent genome-wide analyses of GBM cases have clarified molecular signaling mechanisms underlying GBM biology. However, results of clinical trials targeting phosphorylation-mediated signaling have been unsatisfactory to date. Protein phosphatases are enzymes that antagonize phosphorylation signaling by dephosphorylating phosphorylated signaling molecules. Recently, the critical roles of phosphatases in the regulation of oncogenic signaling in malignant tumor cells have been reported, and tumorigenic roles of deregulated phosphatases have been demonstrated in GBM. However, a detailed mechanism underlying phosphatase-mediated signaling transduction in the regulation of GBM has not been elucidated, and such information is necessary to apply phosphatases as a therapeutic target for GBM. This review highlights and summarizes the phosphatases that have crucial roles in the regulation of oncogenic signaling in GBM cells.
Collapse
|
385
|
Yang J, Nie J, Ma X, Wei Y, Peng Y, Wei X. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer 2019; 18:26. [PMID: 30782187 PMCID: PMC6379961 DOI: 10.1186/s12943-019-0954-x] [Citation(s) in RCA: 1033] [Impact Index Per Article: 172.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/06/2019] [Indexed: 02/07/2023] Open
Abstract
Phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling is one of the most important intracellular pathways, which can be considered as a master regulator for cancer. Enormous efforts have been dedicated to the development of drugs targeting PI3K signaling, many of which are currently employed in clinical trials evaluation, and it is becoming increasingly clear that PI3K inhibitors are effective in inhibiting tumor progression. PI3K inhibitors are subdivided into dual PI3K/mTOR inhibitors, pan-PI3K inhibitors and isoform-specific inhibitors. In this review, we performed a critical review to summarize the role of the PI3K pathway in tumor development, recent PI3K inhibitors development based on clinical trials, and the mechanisms of resistance to PI3K inhibition.
Collapse
Affiliation(s)
- Jing Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ji Nie
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xuelei Ma
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yong Peng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
386
|
Zhao YS, Yang WC, Xin HW, Han JX, Ma SG. MiR-182-5p Knockdown Targeting PTEN Inhibits Cell Proliferation and Invasion of Breast Cancer Cells. Yonsei Med J 2019; 60:148-157. [PMID: 30666836 PMCID: PMC6342713 DOI: 10.3349/ymj.2019.60.2.148] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Breast cancer (BC) is one of the most common malignant tumors, affecting a significant number of women worldwide. MicroRNAs (miRNAs) have been reported to play important roles in tumorigenesis. The aim of this study was to determine the roles of miR-182-5p in BC progression. MATERIALS AND METHODS The expressions of miR-182-5p and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) were measured in BC tissues and cells by quantitative real-time polymerase chain reaction or Western blot. Cell proliferation and invasion were detected by cell counting kit-8 assay and trans-well assay, respectively. The interaction between miR-182-5p and PTEN was probed by bioinformatics analysis, luciferase activity, and RNA immunoprecipitation. A murine xenograft model was established to investigate the role of miR-182-5p in BC progression in vivo. RESULTS An abundance of miR-182-5p was noted in BC tissues and cells. High expression of miR-182-5p was associated with poor survival. Abrogation of miR-182-5p inhibited cell proliferation and invasion in BC cells. Interestingly, PTEN was indicated as a target of miR-182-5p, and its restoration reversed miR-182-5p-mediated promotion of proliferation and invasion of BC cells. Moreover, depletion of miR-182-5p suppressed tumor growth via up-regulating PTEN expression in the murine xenograft model. CONCLUSION MiR-182-5p exhaustion blocked cell proliferation and invasion by regulating PTEN expression, providing a novel therapeutic avenue for treatment of BC.
Collapse
Affiliation(s)
- Yue Sheng Zhao
- Department of Breast Surgery, The Third Hospital Affiliated to Qiqihar Medical College, Qiqihar, China
| | - Wei Chao Yang
- Department of Breast Surgery, Jinan Zhangqiu District Hospital of Traditional Chinese Medicine, Zhangqi District, Jinan, China
| | - Hong Wei Xin
- Department of General Surgery, Sixth People's Hospital of Ji'nan City, Jinan, China
| | - Ji Xia Han
- Department of General Surgery, Sixth People's Hospital of Ji'nan City, Jinan, China
| | - Su Gang Ma
- Department of Breast Surgery, Sixth People's Hospital of Ji'nan City, Jinan, China.
| |
Collapse
|
387
|
Zhang C, Zhou J, Hu J, Lei S, Yuan M, Chen L, Wang G, Qiu Z. Celecoxib attenuates hepatocellular proliferative capacity during hepatocarcinogenesis by modulating a PTEN/NF-κB/PRL-3 pathway. RSC Adv 2019; 9:20624-20632. [PMID: 35515542 PMCID: PMC9065693 DOI: 10.1039/c9ra00429g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/01/2019] [Indexed: 11/21/2022] Open
Abstract
Although the efficacy of celecoxib on various cancer cell behaviors, including aberrant proliferation, in cultured hepatocellular carcinoma (HCC) cells has been demonstrated, whether celecoxib regulates cell proliferation by targeting PRL-3-associated signaling transduction during hepatocarcinogenesis in vivo has been incompletely studied. Here, we investigate the anti-proliferative efficacy of celecoxib in a rapid HCC mouse model established by hydrodynamic transfection of activated AKT and c-Met proto-oncogenes. The results show that celecoxib is effective at delaying the malignant transformation of hepatocytes by reducing the protein expression of Ki67, Cyclin D1 and c-Myc in the AKT/c-Met HCC-bearing mice. Mechanistically, celecoxib increases the protein expression of PTEN and suppresses the protein expression of NF-κB and PRL-3 in the liver of the HCC mice. Using PTEN-silenced and LPS-stimulated approaches in vitro, a mechanism by which celecoxib regulates a PTEN/NF-κB/PRL-3 pathway in HCC cells was illuminated. Altogether, our study demonstrates that celecoxib attenuates the hepatocellular proliferative capacity during hepatocarcinogenesis, which is probably attributable to its regulation of the PTEN/NF-κB/PRL-3 pathway. Celecoxib modulates the PTEN/NF-κB/PRL-3 pathway during hepatocarcinogenesis in vivo.![]()
Collapse
Affiliation(s)
- Cong Zhang
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Junxuan Zhou
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Junjie Hu
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Sheng Lei
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Ming Yuan
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Liang Chen
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
| | - Guihong Wang
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
- Key Laboratory of Chinese Medicine Resource and Compound Prescription
| | - Zhenpeng Qiu
- College of Pharmacy
- Hubei University of Chinese Medicine
- Wuhan
- People's Republic of China
- Key Laboratory of Resources and Chemistry of Chinese Medicine
| |
Collapse
|
388
|
Cai SW, Han Y, Wang GP. miR-148a-3p exhaustion inhibits necrosis by regulating PTEN in acute pancreatitis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5647-5657. [PMID: 31949651 PMCID: PMC6963085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 11/13/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Acute pancreatitis (AP) is a necro-inflammatory disorder with high mortality rate. With advances in understanding the pathogenesis of AP, microRNAs (miRNAs) have been reported to play an essential role in AP progression. However, the mechanism that allows miR-148a-3p to regulate necrosis in AP remains unclear. METHODS Caerulein treatment was used to induce AP in mice or cells. miR-148a-3p-/- mice or miR-148a-3p inhibition in wild type mice were used to investigate the effect of miR-148a-3p on AP. The expression of miR-148a-3p was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The abundances of phosphatase and tensin homolog (PTEN) and hallmarks of necrosis or apoptosis were measured by qRT-PCR or western blots (WB). Cell necrosis, apoptosis, serum amylase or lipase activity and inflammatory cytokines levels were investigated by commercial assay kit. Inflammatory infiltration was analyzed by immunohistochemistry (IHC). The interaction between miR-148a-3p and PTEN was probed by luciferase activity. RESULTS miR-148a-3p was highly expressed in AP and knockout of miR-148a-3p inhibited water content, cell necrosis, amylase and lipase activity while inducing PTEN expression. Moreover, miR-148a-3p deletion attenuated inflammatory infiltration and necrosis by promoting apoptosis. In addition, miR-148a-3p knockdown protected against cell necrosis, amylase, and lipase activity in AP. Intriguingly, PTEN was a target of miR-148a-3p and interference of PTEN reversed the effect of miR-148a-3p deficiency on AP in vitro. CONCLUSION miR-148a-3p inhibition repressed necrosis by regulating PTEN expression in AP, providing a novel biomarker of therapeutics for AP treatment.
Collapse
Affiliation(s)
- Shi-Wen Cai
- Intensive Care Unit, Chang An HospitalXi’an, Shaanxi, China
| | - Ying Han
- Intensive Care Unit, Chang An HospitalXi’an, Shaanxi, China
| | - Guo-Ping Wang
- Emergency Department, Chang An HospitalXi’an, Shaanxi, China
| |
Collapse
|
389
|
Pectolinarigenin inhibits non‑small cell lung cancer progression by regulating the PTEN/PI3K/AKT signaling pathway. Oncol Rep 2018; 40:3458-3468. [PMID: 30542737 PMCID: PMC6196644 DOI: 10.3892/or.2018.6759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is the principal cause of cancer-associated mortality. Pectolinarigenin (Pec) reportedly has effective antitumor activity against certain cancer types. Phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor and serves a vital role in cancer progression. However, the effect of Pec on non-small cell lung cancer (NSCLC) cell proliferation and metastasis, and the underlying mechanism, has not yet been elucidated. In the present study, it was demonstrated that Pec inhibited the proliferation of A549 and Calu-3 cells in dose- and time-dependent manners. The apoptosis rate significantly increased with increasing doses of Pec. Apoptosis-associated protein expression was additionally altered by Pec exposure. Pec was able to suppress the metastasis of NSCLC cells; it upregulated the mRNA and protein expression levels of E-cadherin, and downregulated the mRNA and protein expression levels of vimentin. Additionally, Pec was able to activate PTEN and subsequently downregulate the PI3K/protein kinase B (AKT) signaling pathway. In summary, Pec was able to inhibit cell proliferation, promote apoptosis and suppress metastasis in NSCLC cells through the PTEN/PI3K/AKT signaling pathway, indicating that Pec is a potential agent for NSCLC therapy.
Collapse
|
390
|
Moloughney JG, Vega-Cotto NM, Liu S, Patel C, Kim PK, Wu CC, Albaciete D, Magaway C, Chang A, Rajput S, Su X, Werlen G, Jacinto E. mTORC2 modulates the amplitude and duration of GFAT1 Ser-243 phosphorylation to maintain flux through the hexosamine pathway during starvation. J Biol Chem 2018; 293:16464-16478. [PMID: 30201609 DOI: 10.1074/jbc.ra118.003991] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/28/2018] [Indexed: 12/12/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) controls metabolic pathways in response to nutrients. Recently, we have shown that mTOR complex 2 (mTORC2) modulates the hexosamine biosynthetic pathway (HBP) by promoting the expression of the key enzyme of the HBP, glutamine:fructose-6-phosphate aminotransferase 1 (GFAT1). Here, we found that GFAT1 Ser-243 phosphorylation is also modulated in an mTORC2-dependent manner. In response to glutamine limitation, active mTORC2 prolongs the duration of Ser-243 phosphorylation, albeit at lower amplitude. Blocking glycolysis using 2-deoxyglucose robustly enhances Ser-243 phosphorylation, correlating with heightened mTORC2 activation, increased AMPK activity, and O-GlcNAcylation. However, when 2-deoxyglucose is combined with glutamine deprivation, GFAT1 Ser-243 phosphorylation and mTORC2 activation remain elevated, whereas AMPK activation and O-GlcNAcylation diminish. Phosphorylation at Ser-243 promotes GFAT1 expression and production of GFAT1-generated metabolites including ample production of the HBP end-product, UDP-GlcNAc, despite nutrient starvation. Hence, we propose that the mTORC2-mediated increase in GFAT1 Ser-243 phosphorylation promotes flux through the HBP to maintain production of UDP-GlcNAc when nutrients are limiting. Our findings provide insights on how the HBP is reprogrammed via mTORC2 in nutrient-addicted cancer cells.
Collapse
Affiliation(s)
- Joseph G Moloughney
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Nicole M Vega-Cotto
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Sharon Liu
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Chadni Patel
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Peter K Kim
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Chang-Chih Wu
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Danielle Albaciete
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Cedric Magaway
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Austin Chang
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Swati Rajput
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Xiaoyang Su
- Department of Medicine, Division of Endocrinology, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901
| | - Guy Werlen
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| | - Estela Jacinto
- From the Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854 and
| |
Collapse
|