251
|
Hassing B, Candy A, Eaton CJ, Fernandes TR, Mesarich CH, Di Pietro A, Scott B. Localisation of phosphoinositides in the grass endophyte Epichloë festucae and genetic and functional analysis of key components of their biosynthetic pathway in E. festucae symbiosis and Fusarium oxysporum pathogenesis. Fungal Genet Biol 2022; 159:103669. [PMID: 35114379 DOI: 10.1016/j.fgb.2022.103669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/15/2022] [Accepted: 01/27/2022] [Indexed: 11/24/2022]
Abstract
Phosphoinositides (PI) are essential components of eukaryotic membranes and function in a large number of signaling processes. While lipid second messengers are well studied in mammals and yeast, their role in filamentous fungi is poorly understood. We used fluorescent PI-binding molecular probes to localize the phosphorylated phosphatidylinositol species PI[3]P, PI[3,5]P2, PI[4]P and PI[4,5]P2 in hyphae of the endophyte Epichloë festucae in axenic culture and during interaction with its grass host Lolium perenne. We also analysed the roles of the phosphatidylinositol-4-phosphate 5-kinase MssD and the predicted phosphatidylinositol-3,4,5-triphosphate 3-phosphatase TepA, a homolog of the mammalian tumour suppressor protein PTEN. Deletion of tepA in E. festucae and in the root-infecting tomato pathogen Fusarium oxysporum had no impact on growth in culture or the host interaction phenotype. However, this mutation did enable the detection of PI[3,4,5]P3 in septa and mycelium of E. festucae and showed that TepA is required for chemotropism in F. oxysporum. The identification of PI[3,4,5]P3 in ΔtepA strains suggests that filamentous fungi are able to generate PI[3,4,5]P3 and that fungal PTEN homologs are functional lipid phosphatases. The F. oxysporum chemotropism defect suggests a conserved role of PTEN homologs in chemotaxis across protists, fungi and mammals.
Collapse
Affiliation(s)
- Berit Hassing
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand; Bio-Protection Research Centre, New Zealand
| | - Alyesha Candy
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand; Bio-Protection Research Centre, New Zealand
| | - Carla J Eaton
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand; Bio-Protection Research Centre, New Zealand
| | - Tania R Fernandes
- Departamento de Genética, Campus de Excelencia Internacional Agroalimentario ceiA3, Universidad de Córdoba, Córdoba, Spain
| | - Carl H Mesarich
- Bio-Protection Research Centre, New Zealand; School of Agriculture and Environment, Massey University, Palmerston North, New Zealand
| | - Antonio Di Pietro
- Departamento de Genética, Campus de Excelencia Internacional Agroalimentario ceiA3, Universidad de Córdoba, Córdoba, Spain
| | - Barry Scott
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand; Bio-Protection Research Centre, New Zealand.
| |
Collapse
|
252
|
Novel long non-coding RNA lncAMPC downregulates PTEN via miR-214 to promote nasopharyngeal carcinoma progression. Mol Cell Biochem 2022; 477:805-814. [PMID: 35059924 DOI: 10.1007/s11010-021-04323-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Long non-coding RNAs (lncRNAs) are a type of non-coding RNAs with transcript lengths exceeding 200 nt. lncRNAs suppress or promote cancer mainly by regulating gene expression. The aim of this study was to explore the role of lncRNAs activated in metastatic PCa (lncAMPC) in nasopharyngeal carcinoma (NPC). Total RNAs were isolated from NPC and adjacent non-tumor tissues from 60 NPC patients and subjected to RT-qPCR to analyze differential expression of lncAMPC and miR-214. The roles of lncAMPC and miR-214 in regulating PTEN expression were analyzed using RT-qPCR and Western blot. Cell proliferation was analyzed using the BrdU assay. The results showed that lncAMPC was overexpressed in NPC tissues and was localized in both nuclei and cytoplasms of NPC cells. MiR-214 was positively correlated with lncAMPC. LncAMPC overexpression upregulated miR-214 and indirectly downregulated PTEN via miR-214. BrdU incorporation assay showed that lncAMPC and miR-214 overexpression increased cell proliferation. PTEN overexpression completely reversed the promoting effects of lncAMPC and miR-214 overexpression on cell proliferation. Therefore, lncAMPC might downregulate PTEN via miR-214 to increase NPC cell proliferation.
Collapse
|
253
|
Chowdhury SG, Ray R, Bhattacharya D, Karmakar P. DNA damage induced cellular senescence and it’s PTEN-armed exosomes—the warriors against prostate carcinoma cells. Med Oncol 2022; 39:34. [DOI: 10.1007/s12032-021-01614-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
|
254
|
Feng XH, Wang Y, Wang LY, Shen JF, You CY, Feng QT. Value of PTEN and Echocardiography in Predicting the Efficacy of Trimetazidine Combined with Metoprolol in the Treatment of Heart Failure. Int J Gen Med 2022; 14:10257-10263. [PMID: 34992440 PMCID: PMC8711637 DOI: 10.2147/ijgm.s346538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022] Open
Abstract
Objective To investigate the predictive value of PTEN and echocardiography in the treatment of heart failure with trimetazidine combined with metoprolol. Methods A total of 100 patients with coronary heart disease and HF who admitted to our hospital from August 2018 to August 2020 were enrolled into research. All patients received routine treatment according to the guidelines and were treated with trimetazidine and metoprolol for a total course of 6 months. Echocardiographic parameters and PTEN levels were measured at baseline and after treatment. The patients were divided into groups according to the quartile of basic PTEN level, and the total effective rates were compared. The echocardiographic parameters of patients with different prognosis were analyzed. Bivariate correlation analysis was used to evaluate the correlation between PTEN, echocardiography and treatment effect. Results Compared with that before treatment, the level of PTEN increased significantly after treatment (P < 0.01). According to the quartile of basic PTEN level, the total effective rate of patients with different levels of basic PTEN was was statistically different (P < 0.01). There was a linear correlation between the level of basic PTEN and the treatment effect, and the total effective rate of patients with high level of basic PTEN was higher than that of patients with low level of PTEN. Compared with before treatment, LVEF, SV, E/A and lvfs increased significantly after treatment (P < 0.01). There was a correlation between the basic echocardiographic parameters and the treatment effect of patients. The basic echocardiographic parameters of patients with poor prognosis were worse than those with good prognosis. PTEN expression in patients’ serum was only positively correlated with E/A, but not with LVFE, SV and LVFS (P < 0.01). Conclusion PTEN and echocardiographic parameters serve as a good method to evaluate the short-term therapeutic effect of trimetazidine combined with metoprolol in patients with heart failure.
Collapse
Affiliation(s)
- Xue-Hong Feng
- Department of Echocardiography, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| | - Yang Wang
- Department of Echocardiography, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| | - Lian-Yu Wang
- Department of Echocardiography, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| | - Jun-Fei Shen
- Department of Echocardiography, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| | - Chun-Yuan You
- Department of Echocardiography, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| | - Qiu-Ting Feng
- Department of Cardiology, The Second Hospital of Wuxi Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214002, People's Republic of China
| |
Collapse
|
255
|
Qu H, Gao-Wa H, Hou Y, Ren M, Li J, Jing B, Du Y. TRIM37 interacts with PTEN to promote the growth of human T-cell acute lymphocytic leukemia cells through regulating PI3K/AKT pathway. Front Oncol 2022; 12:1016725. [PMID: 36923153 PMCID: PMC10009101 DOI: 10.3389/fonc.2022.1016725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/30/2022] [Indexed: 03/02/2023] Open
Abstract
Background TRIM37 has been reported to be associated with the tumorigenesis of cancers. However, the role of TRIM37 in T-cell acute lymphoblastic leukemia (T-ALL) remains unclear. This study aimed to characterize the effect of TRIM37 on T-ALL. Methods TRIM37 expression in T-ALL patients and T-ALL cell lines was determined by qRT-PCR and Western blot. Knockdown or overexpression of TRIM37 was conducted by transferring small-interfering TRIM37 or lentivirus-mediated transducing into T-ALL cells. CCK-8 assay and flow cytometry assay were conducted to analyze the proliferation and apoptosis of T-ALL cells. Co-immunoprecipitation experiments were conducted to investigate the relationship between TRIM37 and PTEN and the ubiquitination of PTEN. Results Our results suggested that TRIM37 expression was upregulated in the blood of T-ALL patients and T-ALL cell lines. Knockdown of TRIM37 noticeably inhibited the proliferation and promoted apoptosis of T-ALL cells. Ectopic expression of TRIM37 promoted the proliferation and suppressed the apoptosis rate of MOLT-4 cells and enhanced the phosphorylation of AKT. Moreover, TRIM37 interacted with PTEN and accelerated the degradation of PTEN via TRIM37-mediated ubiquitination in T-ALL cells. Moreover, TRIM37 reduced the sensitivity of T-ALL cells to bortezomib treatment. Additionally, PI3K/AKT signaling pathway was involved in the function of TRIM37 in T-ALL. TRIM37 contributed to the proliferation of T-ALL cells and reduced the susceptibility of T-ALL cells to bortezomib treatment through ubiquitination of PTEN and activating PI3K/AKT signaling pathway. Conclusions Our study suggested that TRIM37 could be considered as a therapeutic target for T-ALL.
Collapse
Affiliation(s)
- Honglan Qu
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - Hasen Gao-Wa
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - Yanyan Hou
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - Mengwei Ren
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - Jun Li
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - Baoshong Jing
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| | - YanDan Du
- Department of Hematology and Oncology, Inner Mongolia Forestry General Hospital, The Second Clinical Medical College of Inner Mongolia University for Nationalities, Yakeshi, China
| |
Collapse
|
256
|
Tayanloo-Beik A, Hamidpour SK, Abedi M, Shojaei H, Tavirani MR, Namazi N, Larijani B, Arjmand B. Zebrafish Modeling of Autism Spectrum Disorders, Current Status and Future Prospective. Front Psychiatry 2022; 13:911770. [PMID: 35911241 PMCID: PMC9329562 DOI: 10.3389/fpsyt.2022.911770] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a complicated range of childhood neurodevelopmental disorders which can occur via genetic or non-genetic factors. Clinically, ASD is associated with problems in relationships, social interactions, and behaviors that pose many challenges for children with ASD and their families. Due to the complexity, heterogeneity, and association of symptoms with some neuropsychiatric disorders such as ADHD, anxiety, and sleep disorders, clinical trials have not yielded reliable results and there still remain challenges in drug discovery and development pipeline for ASD patients. One of the main steps in promoting lead compounds to the suitable drug for commercialization is preclinical animal testing, in which the efficacy and toxicity of candidate drugs are examined in vivo. In recent years, zebrafish have been able to attract the attention of many researchers in the field of neurological disorders such as ASD due to their outstanding features. The presence of orthologous genes for ASD modeling, the anatomical similarities of parts of the brain, and similar neurotransmitter systems between zebrafish and humans are some of the main reasons why scientists draw attention to zebrafish as a prominent animal model in preclinical studies to discover highly effective treatment approaches for the ASD through genetic and non-genetic modeling methods.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamide Shojaei
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nazli Namazi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
257
|
Kim S, Li L, Zhang J, Jiang C, Lü J. Aqueous metabolome of tissue-specific conditional Pten-knockout mouse prostate cancer and TRAMP neuroendocrine carcinoma. Prostate 2022; 82:154-166. [PMID: 34662447 PMCID: PMC9298286 DOI: 10.1002/pros.24256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Metabolic reprograming is now a recognized hallmark of cancer. The prostate-specific phosphatase and tensin homolog deleted on chromosome 10 (Pten) gene-conditional knockout (KO) mouse carcinogenesis model is highly desirable for studying prostate cancer biology and prevention due to its close resemblance of primary molecular defects and histopathological features of human prostate cancer. We have recently published macromolecular profiling of this model by proteomics and transcriptomics, denoting a preeminence of inflammation and myeloid suppressive immune cell features. Here, we performed metabolomic analyses of Pten-KO prostate versus wild type (WT) counterpart for discernable changes in the aqueous metabolites and contrasted to those in the TRAMP neuroendocrine carcinoma (NECa). METHODS Three matched pairs of tissue-specific conditional Pten-KO mouse prostate and WT prostate of litter/cage-mates at 20-22 weeks of age and three pairs of TRAMP NECa versus WT (28-31 weeks) were profiled for their global aqueous metabolite changes, using hydrophilic interaction liquid chromatography-tandem mass spectrometry. RESULTS The Pten-KO prostate increased purine nucleotide pools, cystathionine, and both reduced and oxidized glutathione (GSH, GSSG), and gluconate/glucuronate species in addition to cholesteryl sulfate and polyamine precursor ornithine. On the contrary, Pten-KO prostate contained diminished pools of glycolytic intermediates and phosphorylcholine derivatives, select amino acids, and their metabolites. Bioinformatic integration revealed a significant shunting of glucose away from glycolysis-citrate cycle and glycerol-lipid genesis to pentose phosphate cycle for NADPH/GSH/GSSG redox and pentose moieties for purine and pyrimidine nucleotides, and glycosylation/glucuronidation. Implicit arginine catabolism to ornithine was consistent with immunosuppression in Pten-KO model. While also increased in cystathionine-GSH/GSSG, purine, and pyrimidine nucleotide pools and glucuronidation at the expense of glycolysis-citrate cycle, the TRAMP NECa increased abundance of many amino acids, methyl donor S-adenosyl-methionine, and intermediates for phospholipids without increasing cholesteryl sulfate or ornithine. CONCLUSIONS The aqueous metabolomic patterns in Pten-KO prostate and TRAMP NECa shared similarities in the greater pools of cystathionine, GSH/GSSG redox pair, and nucleotides and shunting away from glycolysis-citrate cycle in both models. Remarkable metabolic distinctions between them included metabolisms of many amino acids (protein synthesis; arginine-ornithine/immune suppression) and cholesteryl sulfate and methylation donor for epigenetic regulations.
Collapse
Affiliation(s)
- Sangyub Kim
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Present address:
Sangyub Kim, Zentalis PharmaceuticalsSan DiegoCaliforniaUSA
| | - Li Li
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
- Present address:
Li Li and Jinhui Zhang, Food and Drug AdministrationSilver SpringMarylandUSA
| | - Jinhui Zhang
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
- Present address:
Li Li and Jinhui Zhang, Food and Drug AdministrationSilver SpringMarylandUSA
| | - Cheng Jiang
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
| | - Junxuan Lü
- Department of PharmacologyPennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Department of Biomedical SciencesTexas Tech University Health Sciences CenterAmarilloTexasUSA
| |
Collapse
|
258
|
Wei X, Hou Y, Long M, Jiang L, Du Y. Molecular mechanisms underlying the role of hypoxia-inducible factor-1 α in metabolic reprogramming in renal fibrosis. Front Endocrinol (Lausanne) 2022; 13:927329. [PMID: 35957825 PMCID: PMC9357883 DOI: 10.3389/fendo.2022.927329] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Renal fibrosis is the result of renal tissue damage and repair response disorders. If fibrosis is not effectively blocked, it causes loss of renal function, leading to chronic renal failure. Metabolic reprogramming, which promotes cell proliferation by regulating cellular energy metabolism, is considered a unique tumor cell marker. The transition from oxidative phosphorylation to aerobic glycolysis is a major feature of renal fibrosis. Hypoxia-inducible factor-1 α (HIF-1α), a vital transcription factor, senses oxygen status, induces adaptive changes in cell metabolism, and plays an important role in renal fibrosis and glucose metabolism. This review focuses on the regulation of proteins related to aerobic glycolysis by HIF-1α and attempts to elucidate the possible regulatory mechanism underlying the effects of HIF-1α on glucose metabolism during renal fibrosis, aiming to provide new ideas for targeted metabolic pathway intervention in renal fibrosis.
Collapse
Affiliation(s)
- Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yue Hou
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Mengtuan Long
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Lili Jiang
- Department of Physical Examination Center, The First Hospital of Jilin University, Changchun, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yujun Du,
| |
Collapse
|
259
|
Khan RIN, Sahu AR, Malla WA, Praharaj MR, Hosamani N, Kumar S, Gupta S, Sharma S, Saxena A, Varshney A, Singh P, Verma V, Kumar P, Singh G, Pandey A, Saxena S, Gandham RK, Tiwari AK. Systems biology under heat stress in Indian cattle. Gene 2021; 805:145908. [PMID: 34411649 DOI: 10.1016/j.gene.2021.145908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 11/26/2022]
Abstract
Transcriptome profiling of Vrindavani and Tharparkar cattle (n = 5 each) revealed that more numbers of genes were dysregulated in Vrindavani than in Tharparkar. A contrast in gene expression was observed with 18.9 % of upregulated genes in Vrindavani downregulated in Tharparkar and 17.8% upregulated genes in Tharparkar downregulated in Vrindavani. Functional annotation of genes differentially expressed in Tharparkar and Vrindavani revealed that the systems biology in Tharparkar is moving towards counteracting the effects due to heat stress. Unlike Vrindavani, Tharparkar is not only endowed with higher expression of the scavengers (UBE2G1, UBE2S, and UBE2H) of misfolded proteins but also with protectors (VCP, Serp1, and CALR) of naïve unfolded proteins. Further, higher expression of the antioxidants in Tharparkar enables it to cope up with higher levels of free radicals generated as a result of heat stress. In this study, we found relevant genes dysregulated in Tharparkar in the direction that can counter heat stress.
Collapse
Affiliation(s)
- Raja Ishaq Nabi Khan
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Amit Ranjan Sahu
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Waseem Akram Malla
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Manas Ranjan Praharaj
- Computational Biology and Genomics, National Institute of Animal Biotechnology, Hyderabad, India
| | - Neelima Hosamani
- Computational Biology and Genomics, National Institute of Animal Biotechnology, Hyderabad, India
| | - Shakti Kumar
- Computational Biology and Genomics, National Institute of Animal Biotechnology, Hyderabad, India
| | - Smita Gupta
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Shweta Sharma
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Archana Saxena
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Anshul Varshney
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Pragya Singh
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Vinay Verma
- Division of Physiology and Climatology, Indian Veterinary Research Institute, Bareilly, India
| | - Puneet Kumar
- Division of Physiology and Climatology, Indian Veterinary Research Institute, Bareilly, India
| | - Gyanendra Singh
- Division of Physiology and Climatology, Indian Veterinary Research Institute, Bareilly, India
| | - Aruna Pandey
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Shikha Saxena
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly, India
| | - Ravi Kumar Gandham
- Computational Biology and Genomics, National Institute of Animal Biotechnology, Hyderabad, India.
| | - Ashok Kumar Tiwari
- Division of Biological Standardization, Indian Veterinary Research Institute, Bareilly, India.
| |
Collapse
|
260
|
Zhai Z, Mu T, Zhao L, Li Y, Zhu D, Pan Y. MiR-181a-5p facilitates proliferation, invasion, and glycolysis of breast cancer through NDRG2-mediated activation of PTEN/AKT pathway. Bioengineered 2021; 13:83-95. [PMID: 34951340 PMCID: PMC8805873 DOI: 10.1080/21655979.2021.2006974] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of microRNAs (miRNAs) is associated with the occurrence and development of breast cancer. In this research, we explored the involvement of miR-181a-5p in the progression of breast cancer and investigated potential molecular mechanisms. Firstly, the miR-181a-5p and N-myc downstream-regulated gene (NDRG) 2 expression was detected by real-time quantitative polymerase chain reaction. Cellular processes were assessed using Cell Counting Kit 8, Bromodeoxyuridine, colony formation and transwell assays. HK2, PKM2 and LDHA activities were assessed by ELISA. The combination between miR-181a-5p was assessed by dual-luciferase reporter assay and RNA pull-down assay. The results indicated that miR-181a-5p levels were upregulated and NDRG2 levels were downregulated in breast cancer, leading to poor prognosis. Silencing of miR-181a-5p inhibited cell proliferation, invasion, glycolysis, and xenograft tumor growth, while enhanced miR-181a-5p got the opposite results. Furthermore, NDRG2 acts as a target of miR-181a-5p. Knockout of NDRG2 facilitated biological behaviors and meanwhile enhanced phosphorylation (p)-PTEN and p-AKT levels. Rescue experiments showed that restoring NDRG2 abolished the effects caused by miR-181a-5p in breast cancer cells. In conclusion, miR-181a-5p facilitated tumor progression through NDRG2-induced activation of PTEN/AKT signaling pathway of breast cancer, suggesting that focusing on miR-181a-5p may provide new insight for breast cancer therapy. Abbreviations Brdu: Bromodeoxyuridine; CCK-8: Cell Counting Kit-8; miRNA: microRNAs; mut: mutant; RT-qPCR: real-time quantitative polymerase chain reaction; UTR: untranslated region; WT: wild-type
Collapse
Affiliation(s)
- Zhen Zhai
- Breast Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Tianlong Mu
- Breast Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
- Pathology Department, Dongfang Hostipal Beijing University of Chinese Medicine, Beijing, China
| | - Lina Zhao
- Breast Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yiliang Li
- Breast Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Dongsheng Zhu
- Breast Department, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yanshu Pan
- Periodical Center, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
261
|
Tailor K, Paul J, Ghosh S, Kumari N, Kwabi-Addo B. RASAL2 suppresses the proliferative and invasive ability of PC3 prostate cancer cells. Oncotarget 2021; 12:2489-2499. [PMID: 34966481 PMCID: PMC8711570 DOI: 10.18632/oncotarget.28158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/08/2021] [Indexed: 11/25/2022] Open
Abstract
The RAS protein activator like 2 (RASAL2) negatively regulates RAS proto-oncogene which is activated by high mutation rate in cancer. Thus, RASAL2 expression could potentially limit the function of RAS in prostate cancer (PCa). Genome-wide DNA methylation analysis demonstrated that RASAL2 is differentially hypermethylated in PCa tissues compared to benign prostate tissues. The PCR analysis of RASAL2 mRNA transcript showed differential expression in a panel of prostate cell lines with most PCa showing lower RASAL2 expression compared to benign prostatic epithelial cells. In PCa PC3 cells, the ectopic expression of RASAL2 significantly inhibited cell proliferation and invasion and induced an S phase plus G2/M phase cell cycle arrest. Ingenuity Pathway Analysis (IPA) demonstrated a cross talk between RASAL2 and TNFα, a key cytokine in immune signaling pathway that is relevant in PCa. Over-expression of RASAL2 downregulated TNFα expression whereas the knockdown of RASAL2 caused increased expression of TNFα. Taken together, our data demonstrates tumor suppressor role for RASAL2 in human PCa cells, despite increased RAS oncogenic activity. Our observation provides a new mechanistic insight of RASAL2 expression in aberrant Ras expression and immune signaling in PCa cells suggesting a potential novel therapeutic target for PCa.
Collapse
Affiliation(s)
- Krishma Tailor
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA
| | - Joseph Paul
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA
| | - Somiranjan Ghosh
- 2Department of Biology, Howard University, Washington, DC 20059, USA
| | - Namita Kumari
- 3Center for Sickle Cell Disease, Howard University, Washington, DC 20059, USA
| | - Bernard Kwabi-Addo
- 1Department of Biochemistry and Molecular Biology, Howard University, Washington, DC 20059, USA,Correspondence to:Bernard Kwabi-Addo, email:
| |
Collapse
|
262
|
Wang Q, Wang J, Xiang H, Ding P, Wu T, Ji G. The biochemical and clinical implications of phosphatase and tensin homolog deleted on chromosome ten in different cancers. Am J Cancer Res 2021; 11:5833-5855. [PMID: 35018228 PMCID: PMC8727805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/08/2021] [Indexed: 06/14/2023] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome ten (PTEN) is widely known as a tumor suppressor gene. It is located on chromosome 10q23 with 200 kb, and has dual activity of both protein and lipid phosphatase. In addition, as a targeted gene in multiple pathways, PTEN has a variety of physiological activities, such as those regulating the cell cycle, inducing cell apoptosis, and inhibiting cell invasion, etc. The PTEN gene have been identified in many kinds of cancers due to its mutations, deletions and inactivation, such as lung cancer, liver cancer, and breast cancer, and they are closely connected with the genesis and progression of cancers. To a large extent, the tumor suppressive function of PTEN is realized through its inhibition of the PI3K/AKT signaling pathway which controls cells apoptosis and development. In addition, PTEN loss has been associated with the prognosis of many cancers, such as lung cancer, liver cancer, and breast cancer. PTEN gene is related to many cancers and their pathological development. On the basis of a large number of related studies, this study describes in detail the structure, regulation, function and classical signal pathways of PTEN, as well as the relationship between various tumors related to PTEN. In addition, some drug studies targeting PTEN/PI3K/AKT/mTOR are also introduced in order to provide some directions for experimental research and clinical treatment of tumors.
Collapse
Affiliation(s)
- Qinyi Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Peilun Ding
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| |
Collapse
|
263
|
Ghafarkhani M, Avci CB, Rahbarghazi R, Karimi A, Sadeghizadeh M, Zarebkohan A, Bani F. Mild hyperthermia induced by gold nanorods acts as a dual-edge blade in the fate of SH-SY5Y cells via autophagy. Sci Rep 2021; 11:23984. [PMID: 34907215 PMCID: PMC8671444 DOI: 10.1038/s41598-021-02697-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Unraveling unwanted side effects of nanotechnology-based therapies like photothermal therapy (PTT) is vital in translational nanomedicine. Herein, we monitored the relationship between autophagic response at the transcriptional level by using a PCR array and tumor formation ability by colony formation assay in the human neuroblastoma cell line, SH-SY5Y, 48 h after being exposed to two different mild hyperthermia (43 and 48 °C) induced by PTT. In this regard, the promotion of apoptosis and autophagy were evaluated using immunofluorescence imaging and flow cytometry analyses. Protein levels of Ki-67, P62, and LC3 were measured using ELISA. Our results showed that of 86 genes associated with autophagy, the expression of 54 genes was changed in response to PTT. Also, we showed that chaperone-mediated autophagy (CMA) and macroautophagy are stimulated in PTT. Importantly, the results of this study also showed significant changes in genes related to the crosstalk between autophagy, dormancy, and metastatic activity of treated cells. Our findings illustrated that PTT enhances the aggressiveness of cancer cells at 43 °C, in contrast to 48 °C by the regulation of autophagy-dependent manner.
Collapse
Affiliation(s)
- Maryam Ghafarkhani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 516661-4733, Tabriz, Iran
| | - Cigir Biray Avci
- Department of Medical Biology, Medical Faculty, Ege University, Bornova, 35100, Izmir, Turkey
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Sadeghizadeh
- Department of Nanobiotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 516661-4733, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Farhad Bani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 516661-4733, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
264
|
Zhou L, Ouyang T, Li M, Hong T, Mhs A, Meng W, Zhang N. Ubiquitin-Specific Peptidase 7: A Novel Deubiquitinase That Regulates Protein Homeostasis and Cancers. Front Oncol 2021; 11:784672. [PMID: 34869041 PMCID: PMC8640129 DOI: 10.3389/fonc.2021.784672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/26/2021] [Indexed: 12/23/2022] Open
Abstract
Ubiquitin-Specific Peptidase 7 (USP7), or herpes virus-associated protease (HAUSP), is the largest family of the deubiquitinating enzymes (DUBs). Recent studies have shown that USP7 plays a vital role in regulating various physiological and pathological processes. Dysregulation of these processes mediated by USP7 may contribute to many diseases, such as cancers. Moreover, USP7 with aberrant expression levels and abnormal activity are found in cancers. Therefore, given the association between USP7 and cancers, targeting USP7 could be considered as an attractive and potential therapeutic approach in cancer treatment. This review describes the functions of USP7 and the regulatory mechanisms of its expression and activity, aiming to emphasize the necessity of research on USP7, and provide a better understanding of USP7-related biological processes and cancer.
Collapse
Affiliation(s)
- Lin Zhou
- First Clinical Medical College, Nanchang University, Nanchang, China
| | - Taohui Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Alriashy Mhs
- Department of Neurosurgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Wei Meng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Na Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
265
|
Sasidharakurup H, Kumar G, Nair B, Diwakar S. Mathematical Modeling of Severe Acute Respiratory Syndrome Coronavirus 2 Infection Network with Cytokine Storm, Oxidative Stress, Thrombosis, Insulin Resistance, and Nitric Oxide Pathways. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:770-781. [PMID: 34807729 DOI: 10.1089/omi.2021.0155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a systemic disease affecting not only the lungs but also multiple organ systems. Clinical studies implicate that SARS-CoV-2 infection causes imbalance of cellular homeostasis and immune response that trigger cytokine storm, oxidative stress, thrombosis, and insulin resistance. Mathematical modeling can offer in-depth understanding of the SARS-CoV-2 infection and illuminate how subcellular mechanisms and feedback loops underpin disease progression and multiorgan failure. We report here a mathematical model of SARS-CoV-2 infection pathway network with cytokine storm, oxidative stress, thrombosis, insulin resistance, and nitric oxide (NO) pathways. The biochemical systems theory model shows autocrine loops with positive feedback enabling excessive immune response, cytokines, transcription factors, and interferons, which can imbalance homeostasis of the system. The simulations suggest that changes in immune response led to uncontrolled release of cytokines and chemokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor α (TNFα), and affect insulin, coagulation, and NO signaling pathways. Increased production of NETs (neutrophil extracellular traps), thrombin, PAI-1 (plasminogen activator inhibitor-1), and other procoagulant factors led to thrombosis. By analyzing complex biochemical reactions, this model forecasts the key intermediates, potential biomarkers, and risk factors at different stages of COVID-19. These insights can be useful for drug discovery and development, as well as precision treatment of multiorgan implications of COVID-19 as seen in systems medicine.
Collapse
Affiliation(s)
- Hemalatha Sasidharakurup
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Geetha Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Bipin Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Shyam Diwakar
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- School of Engineering, Amrita Vishwa Vidyapeetham, Kollam, India
| |
Collapse
|
266
|
Peng P, Yu H, Xing C, Tao B, Li C, Huang J, Ning G, Zhang B, Feng S. Exosomes-mediated phenotypic switch of macrophages in the immune microenvironment after spinal cord injury. Biomed Pharmacother 2021; 144:112311. [PMID: 34653754 DOI: 10.1016/j.biopha.2021.112311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/25/2021] [Accepted: 10/05/2021] [Indexed: 02/08/2023] Open
Abstract
Although accumulating evidence indicated that modulating macrophage polarization could ameliorate the immune microenvironment and facilitate the repair of spinal cord injury (SCI), the underlying mechanism of macrophage phenotypic switch is still poorly understood. Exosomes (Exos), a potential tool of cell-to-cell communication, may play important roles in cell reprogramming. Herein, we investigated the roles of macrophages-derived exosomes played for macrophage polarization in the SCI immune microenvironment. In this study, we found the fraction of M2 macrophages was markedly decreased after SCI. Moreover, the M2 macrophages-derived exosomes could increase the percentage of M2 macrophages, decrease that of M1 macrophages while the M1 macrophages-derived exosomes acted oppositely. According to the results of in silico analyses and molecular experiments verification, this phenotypic switch might be mediated by the exosomal miRNA-mRNA network, in which the miR-23a-3p/PTEN/PI3K/AKT axis might play an important role. In conclusion, our study suggests macrophage polarization that regulated by various interventions might be mediated by their own exosomes at last. Moreover, M2 macrophages-derived exosomes could promote M2 macrophage polarization via the potential miRNA-mRNA network. Considering its potential of modulating polarization, M2 macrophages-derived exosomes may be a promising therapeutic agent for SCI repair.
Collapse
Affiliation(s)
- Peng Peng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Yu
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cong Xing
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Tao
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Li
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingyuan Huang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Guangzhi Ning
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Zhang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
267
|
Beneficial Oxidative Stress-Related trans-Resveratrol Effects in the Treatment and Prevention of Breast Cancer. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112211041] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resveratrol is one of the most investigated polyphenols for its multiple biological activities and many beneficial effects. These are mainly related to its ability to scavenge free radicals and reduce oxidative stress. Resveratrol has also been shown to have the ability to stimulate the production of antioxidant enzymes, which interact with numerous signaling pathways involved in tumor development, and to possess side effects associated with the use of chemotherapy drugs. In this review article we summarized the main discoveries about the impact resveratrol can have in helping to prevent, as well as adjuvant treating, breast cancer. A brief overview of the primary sources of resveratrol as well as some approaches for improving its bioavailability have been also discussed.
Collapse
|
268
|
Zughaibi TA, Suhail M, Tarique M, Tabrez S. Targeting PI3K/Akt/mTOR Pathway by Different Flavonoids: A Cancer Chemopreventive Approach. Int J Mol Sci 2021; 22:12455. [PMID: 34830339 PMCID: PMC8621356 DOI: 10.3390/ijms222212455] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is, globally, one of the main causes of death. Even though various therapies are available, they are still painful because of their adverse side effects. Available treatments frequently fail due to unpromising responses, resistance to classical anticancer drugs, radiation therapy, chemotherapy, and low accessibility to tumor tissues. Developing novel strategies to minimize adverse side effects, improve chemotherapy sensitivity, and control cancer progression is needed. Many studies have suggested small dietary molecules as complementary treatments for cancer patients. Different components of herbal/edible plants, known as flavonoids, have recently garnered attention due to their broad biological properties (e.g., antioxidant, antiviral, antimicrobial, anti-inflammatory, anti-mutagenic, anticancer, hepatoprotective, and cardioprotective). These flavonoids have shown anticancer activity by affecting different signaling cascades. This article summarizes the key progress made in this area and discusses the role of flavonoids by specifically inhibiting the PI3K/Akt/mTOR pathway in various cancers.
Collapse
Affiliation(s)
- Torki A. Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Tarique
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO 65201, USA;
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
269
|
Rivas DA, Peng F, Benard T, Ramos da Silva AS, Fielding RA, Margolis LM. miR-19b-3p is associated with a diametric response to resistance exercise in older adults and regulates skeletal muscle anabolism via PTEN inhibition. Am J Physiol Cell Physiol 2021; 321:C977-C991. [PMID: 34705586 PMCID: PMC8714992 DOI: 10.1152/ajpcell.00190.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Understanding paradoxical responses to anabolic stimulation and identifying the mechanisms for this inconsistency in mobility-limited older adults may provide new targets for the treatment of sarcopenia. Our laboratory has discovered that dysregulation in microRNA (miRNA) that target anabolic pathways is a potential mechanism resulting in age-associated decreases in skeletal muscle mass and function (sarcopenia). The objective of the current study was to assess circulating miRNA expression profiles in diametric response of leg lean mass in mobility-limited older individuals after a 6-mo progressive resistance exercise training intervention (PRET) and determine the influence of differentially expressing miRNA on regulation of skeletal muscle mass. Participants were dichotomized by gain (Gainers; mean +561.4 g, n = 33) or loss (Losers; mean −589.8 g, n = 40) of leg lean mass after PRET. Gainers significantly increased fat-free mass 2.4% vs. −0.4% for Losers. Six miRNA (miR-1-3p, miR-19b-3p, miR-92a, miR-126, miR-133a-3p, and miR-133b) were significantly identified to be differentially expressed between Gainers and Losers, with miR-19b-3p being the miRNA most highly associated with increases in fat-free mass. Using an aging mouse model, we then assessed if miR-19b-3p expression was different in young mice with larger muscle mass compared with older mice. Circulating and skeletal muscle miR-19b-3p expression was higher in young compared with old mice and was positively associated with muscle mass and grip strength. We then used a novel integrative approach to determine if differences in circulating miR-19b-3p potentially translate to augmented anabolic response in human skeletal muscle cells in vitro. Results from this analysis identified that overexpression of miR-19b-3p targeted and downregulated PTEN by 64% to facilitate significant ∼50% increase in muscle protein synthetic rate as measured with SUnSET. The combine results of these three models identify miR-19b-3p as a potent regulator of muscle anabolism that may contribute to an inter-individual response to PRET in mobility-limited older adults.
Collapse
Affiliation(s)
- Donato A Rivas
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Fei Peng
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Townsend Benard
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Adelino Sanchez Ramos da Silva
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,School of Physical Education and Sport of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Roger A Fielding
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Lee M Margolis
- Nutrition, Exercise Physiology and Sarcopenia Laboratory; Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.,Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States
| |
Collapse
|
270
|
Ari Yuka S, Yilmaz A. Effect of SARS-CoV-2 infection on host competing endogenous RNA and miRNA network. PeerJ 2021; 9:e12370. [PMID: 34722003 PMCID: PMC8541317 DOI: 10.7717/peerj.12370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022] Open
Abstract
Competing endogenous RNAs (ceRNA) play a crucial role in cell functions. Computational methods that provide large-scale analysis of the interactions between miRNAs and their competitive targets can contribute to the understanding of ceRNA regulations and critical regulatory functions. Recent reports showed that viral RNAs can compete with host RNAs against host miRNAs. Regarding SARS-CoV-2 RNA, no comprehensive study had been reported about its competition with cellular ceRNAs. In this study, for the first time, we used the ceRNAnetsim package to assess ceRNA network effects per individual cell and competitive behavior of SARS-CoV-2 RNA in the infected cells using single-cell sequencing data. Our computations identified 195 genes and 29 miRNAs which vary in competitive behavior specifically in presence of SARS-CoV-2 RNA. We also investigated 18 genes that are affected by genes that lost perturbation ability in presence of SARS-CoV-2 RNA in the human miRNA:ceRNA network. These transcripts have associations with COVID-19-related symptoms as well as many dysfunctions such as metabolic diseases, carcinomas, heart failure. Our results showed that the effects of the SARS-CoV-2 genome on host ceRNA interactions and consequent dysfunctions can be explained by competition among various miRNA targets. Our perturbation ability perspective has the potential to reveal yet to be discovered SARS-CoV-2 induced effects invisible to conventional approaches.
Collapse
Affiliation(s)
- Selcen Ari Yuka
- Department of Bioengineering, Yildiz Technical University, Istanbul, Turkey
| | - Alper Yilmaz
- Department of Bioengineering, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
271
|
The Effect of miR-520b on Macrophage Polarization and T Cell Immunity by Targeting PTEN in Breast Cancer. JOURNAL OF ONCOLOGY 2021; 2021:5170496. [PMID: 34659411 PMCID: PMC8514911 DOI: 10.1155/2021/5170496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/31/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Background Breast cancer is the most common cancer in women. miR-520b had binding sites with PTEN through the bioinformatics prediction. But few studies have been conducted on miR-520b and PTEN in breast cancer. We aimed to explore the effect of miR-520b and PTEN on breast cancer and the mechanisms involved. Methods Clinical samples of breast cancer were collected. Bioinformatics analysis was performed to screen the differentially expressed miRNAs. CD4 T cells and CD8 T cells were cocultured with MCF-7 cells in the Transwell system. Moreover, MCF-7 cells and M0 macrophage cocultured cell lines were constructed. qRT-PCR, IF, western blot, flow cytometry, and ELISA were performed to detect related factors expression. Starbase and dual-luciferase reporter assay verified the binding of miR-520b to PTEN. The tumor formation model was established to study miR-520b and PTEN effects in vivo. Results The differentially expressed miR-520b was screened via miRNAs sequencing and cell verification. miR-520b expression was high, PTEN was low in tumor tissues, T cells and NK cells were inhibited, and macrophages were transformed into M2 type, promoting immune escape. In addition, miR-520b bound to PTEN. Then, splenic CD4 T cells and CD8 T cells were successfully sorted. During CD4 T cell differentiation to Th1 and Treg, Th1 was inhibited, and Treg was activated. We found the polarization of macrophages was related to breast cancer. The proportion of CD206 cells increased and CD68 cells decreased in the miR-520b mimics group compared with the mimic NC group. Compared with the inhibitor NC group, the proportion of CD206 cells decreased, and CD68 cells increased in the miR-520b inhibitor group. In vivo experiments showed that miR-520b inhibitor inhibited tumor growth and promoted PTEN expression. The proportion of CD3, CD4, CD8, NK1.1, CD4+IFNγ, and CD68 cells increased, while FOXP3 and CD206 cells decreased in the miR-520b inhibitor group compared with the inhibitor NC group. However, the proportion of CD3, CD4, CD8, NK1.1, CD4+IFNγ, and CD68 cells decreased, while FOXP3 and CD206 cells increased after the addition of siPTEN. Conclusions miR-520b inhibited PTEN and aggravated breast tumors. miR-520b inhibitor enhanced CD4 and CD8 cell populations in the tumor immune microenvironment and inhibited tumor growth.
Collapse
|
272
|
Xin L, Zhou LQ, Liu C, Zeng F, Yuan YW, Zhou Q, Li SH, Wu Y, Wang JL, Wu DZ, Lu H. Transfer of LncRNA CRNDE in TAM-derived exosomes is linked with cisplatin resistance in gastric cancer. EMBO Rep 2021; 22:e52124. [PMID: 34647680 DOI: 10.15252/embr.202052124] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
This study explores the role of the long noncoding RNA (LncRNA) CRNDE in cisplatin (CDDP) resistance of gastric cancer (GC) cells. Here, we show that LncRNA CRNDE is upregulated in carcinoma tissues and tumor-associated macrophages (TAMs) of GC patients. In vitro experiments show that CRNDE is enriched in M2-polarized macrophage-derived exosomes (M2-exo) and is transferred from M2 macrophages to GC cells via exosomes. Silencing CRNDE in M2-exo reverses the promotional effect of M2-exo on cell proliferation in CDDP-treated GC cells and homograft tumor growth in CDDP-treated nude mice. Mechanistically, CRNDE facilitates neural precursor cell expressed developmentally downregulated protein 4-1 (NEDD4-1)-mediated phosphatase and tensin homolog (PTEN) ubiquitination. Silencing CRNDE in M2-exo enhances the CDDP sensitivity of GC cells treated with M2-exo, which is reduced by PTEN knockdown. Collectively, these data reveal a vital role for CRNDE in CDDP resistance of GC cells and suggest that the upregulation of CRNDE in GC cells may be attributed to the transfer of TAM-derived exosomes.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li-Qiang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chuan Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Zeng
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi-Wu Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shi-Hao Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - You Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin-Liang Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Deng-Zhong Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hao Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
273
|
Zhang Y, Zheng A. MiR-142-5p promotes retinoblastoma cell proliferation, migration and invasion by targeting PTEN. J Biochem 2021; 170:195-202. [PMID: 34562091 DOI: 10.1093/jb/mvaa121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/11/2020] [Indexed: 11/13/2022] Open
Abstract
The study intends to probe the functions of miR-142-5p in retinoblastoma (RB) and the relationship between miR-142-5p and phosphatase and tensin homolog deleted on chromosome ten (PTEN). In our study, miR-142-5p and PTEN mRNA expression in RB tissue, serum of RB patients and RB cell lines were investigated by quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation, migration, invasion and cell apoptosis were measured using MTT assay, BrdU assay, Transwell experiments and flow cytometry analysis, respectively. Binding sites between miR-142-5p and PTEN were predicted by the TargetScan database and were confirmed via qRT-PCR, western blot and dual-luciferase reporter gene assay. It was demonstrated that miR-142-5p expression was elevated in RB tissue, serum of RB patients and RB cell lines. MiR-142-5p overexpression remarkably promoted the proliferation, migration, invasion and inhibited the apoptosis of WERI-RB-1 cells while miR-142-5p knockdown induced opposite effects in Y79 cells. MiR-142-5p decreased PTEN expression in both mRNA and protein expression levels, and PTEN was identified as a target gene of miR-142-5p. Cotransfection of PTEN overexpression plasmids reversed the influences of miR-142-5p on RB cells. In conclusion, miR-142-5p enhances proliferation, migration and invasion of RB cell by targeting PTEN.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Ophthalmology, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Ailing Zheng
- Department of Ophthalmology, Shanxian Central Hospital, Wenhua Road No. 1, Heze 274300, Shandong, China
| |
Collapse
|
274
|
Xu S, Wang T, Lu X, Zhang H, Liu L, Kong X, Li S, Wang X, Gao H, Wang J, Wang L. Identification of LINC00173 in Myasthenia Gravis by Integration Analysis of Aberrantly Methylated- Differentially Expressed Genes and ceRNA Networks. Front Genet 2021; 12:726751. [PMID: 34603387 PMCID: PMC8481885 DOI: 10.3389/fgene.2021.726751] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease associated with autoantibody production that leads to skeletal muscle weakness. The molecular mechanisms underlying MG are not fully understood. We analyzed the gene expression profile (GSE85452) and methylation profile (GSE85647) of MG samples from the GEO database to identify aberrantly methylated-differentially expressed genes. By integrating the datasets, we identified 143 hypermethylation-low expression genes and 91 hypomethylation-high expression genes. Then we constructed PPI network and ceRNA networks by these genes. Phosphatase and tensin homolog (PTEN) and Abelson tyrosine-protein kinase (ABL)1 were critical genes in both PPI networks and ceRNA networks. And potential MG associated lncRNAs were selected by comprehensive analysis of the critical genes and ceRNA networks. In the hypermethylation-low expression genes associated ceRNA network, sirtuin (SIRT)1 was the most important gene and the lncRNA HLA complex (HC) P5 had the highest connection degree. Meanwhile, PTEN was the most important gene and the lncRNA LINC00173 had the highest connection degree in the hypomethylation-high expression genes associated ceRNA network. LINC00173 was validated to be upregulated in MG patients by qRT-PCR (P = 0.005), which indicated LINC00173 might be a potential biomarker for MG. These results provide a basis for future studies on the molecular pathogenesis of MG.
Collapse
Affiliation(s)
- Si Xu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianfeng Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Lu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Liu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaotong Kong
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuang Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongyu Gao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
275
|
Wolf J, Dong C, O'Day EM. Metabolite Biomarkers of Response (BoRs): Towards a fingerprint for the evolution of metastatic breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 165:8-18. [PMID: 34419530 DOI: 10.1016/j.pbiomolbio.2021.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
Breast cancer is the most common cancer in women worldwide and despite improved treatment strategies, it persists as the second leading cause of death of women globally. Overall prognosis drops drastically once the cancer has metastasized, which is also associated with resistance to therapy. The evolution from a localized breast cancer to metastatic disease is complex and multifactorial. Metabolic reprogramming is a pre-requisite for this transition. In this graphical review, we provide an overview of altered metabolic pathways observed in metastatic breast cancer (mBC) and detail how metabolite biomarkers could serve as a novel class of precision medicine tools to improve the diagnosis, monitoring, and treatment of mBC.
Collapse
Affiliation(s)
| | - Chen Dong
- Olaris, Inc, Waltham, MA, 02451, USA
| | | |
Collapse
|
276
|
Xia RM, Liu T, Li WG, Xu XQ. RNA-binding protein RBM24 represses colorectal tumourigenesis by stabilising PTEN mRNA. Clin Transl Med 2021; 11:e383. [PMID: 34709758 PMCID: PMC8506628 DOI: 10.1002/ctm2.383] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND RNA-binding motif protein 24 (RBM24) functions as a splicing regulator, which is critical for organ development and is dysregulated in human cancers. Here, we aim to uncover the biological function of RBM24 in colorectal tumourigenesis. METHODS Xenograft tumour model, Rbm24 knockout and Apcmin/+ mouse models were utilised. Colorectal cancer cells overexpressing or silencing RBM24 were established. RNA immunoprecipitation (RIP) assay was conducted to detect protein-RNA associations. Gene expression was measured by immunohistochemistry, western blotting, or quantitative PCR (qPCR). RESULTS Rbm24-knockout mice developed spontaneous colorectal adenomas with lower expression of phosphatase and tensin homolog (PTEN). Immunohistochemical staining for the proliferation markers Ki-67 and pHH3 and BrdU assay showed intestinal hyperplasia in Rbm24-knockout mice compared to wild-type mice. RBM24 expression in colorectal adenoma tissues of Apcmin/+ mouse was downregulated compared with adjacent normal samples and was positively correlated with PTEN expression. In vitro, RBM24 overexpression suppressed cell proliferation, migration, invasion and increased sensitivity to 5-FU or cisplatin in CRC cells. Mechanistically, RBM24 maintained PTEN mRNA stability by directly binding to the GT-rich region at positions 8101-8251 in the 3'-UTR of PTEN mRNA, prolonging the half-life of PTEN mRNA, thereby increasing PTEN expression. Hence, low expression of RBM24 downregulated PTEN mRNA, causing the activation of PI3K-Akt signalling in CRC cells. Furthermore, RBM24 expression in CRC tissues was lower than adjacent normal samples. RBM24 expression was positively correlated with PTEN expression and negatively correlated with Ki-67 level. CRC patients with high RBM24 expression had a favourable outcome. CONCLUSIONS Taken together, RBM24 expression is markedly lower in colorectal tumours than in para-carcinoma tissues. Rbm24-knockout mice develop spontaneous colorectal adenomas. RBM24 directly binds and stabilises PTEN mRNA, which could cause the suppression of CRC cell proliferation, migration and invasion, thereby repressing colorectal tumourigenesis. These findings support the tumour-suppressive role of RBM24. Targeting RBM24 holds strong promise for the diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Rong Mu Xia
- Institute of Stem Cell and Regenerative MedicineSchool of MedicineXiamen UniversityXiamenFujianPeople's Republic of China
| | - Tao Liu
- Department of Hepatobiliary SurgerySchool of MedicineXiang'an Hospital of Xiamen UniversityXiamen UniversityXiamenFujianPeople's Republic of China
| | - Wen Gang Li
- Institute of Stem Cell and Regenerative MedicineSchool of MedicineXiamen UniversityXiamenFujianPeople's Republic of China
- Department of Hepatobiliary SurgerySchool of MedicineXiang'an Hospital of Xiamen UniversityXiamen UniversityXiamenFujianPeople's Republic of China
| | - Xiu Qin Xu
- Institute of Stem Cell and Regenerative MedicineSchool of MedicineXiamen UniversityXiamenFujianPeople's Republic of China
| |
Collapse
|
277
|
Etiologies of Melanoma Development and Prevention Measures: A Review of the Current Evidence. Cancers (Basel) 2021; 13:cancers13194914. [PMID: 34638397 PMCID: PMC8508267 DOI: 10.3390/cancers13194914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Melanoma constitutes a major public health risk, with the rates of diagnosis increasing on a yearly basis. Monitoring for risk factors and preventing dangerous behaviors that increase melanoma risk, such as tanning, are important measures for melanoma prevention. Additionally, assessing the effectiveness of various methods to prevent sun exposure and sunburns—which can lead to melanoma—is important to help identify ways to reduce the development of melanoma. We summarize the recent evidence regarding the heritable and behavioral risks underlying melanoma, as well as the current methods used to reduce the risk of developing melanoma and to improve the diagnosis of this disease. Abstract (1) Melanoma is the most aggressive dermatologic malignancy, with an estimated 106,110 new cases to be diagnosed in 2021. The annual incidence rates continue to climb, which underscores the critical importance of improving the methods to prevent this disease. The interventions to assist with melanoma prevention vary and typically include measures such as UV avoidance and the use of protective clothing, sunscreen, and other chemopreventive agents. However, the evidence is mixed surrounding the use of these and other interventions. This review discusses the heritable etiologies underlying melanoma development before delving into the data surrounding the preventive methods highlighted above. (2) A comprehensive literature review was performed to identify the clinical trials, observational studies, and meta-analyses pertinent to melanoma prevention and incidence. Online resources were queried to identify epidemiologic and clinical trial information. (3) Evidence exists to support population-wide screening programs, the proper use of sunscreen, and community-targeted measures in the prevention of melanoma. Clinical evidence for the majority of the proposed preventive chemotherapeutics is presently minimal but continues to evolve. (4) Further study of these chemotherapeutics, as well as improvement of techniques in artificial intelligence and imaging techniques for melanoma screening, is warranted for continued improvement of melanoma prevention.
Collapse
|
278
|
Pomar CA, Serra F, Palou A, Sánchez J. Lower miR-26a levels in breastmilk affect gene expression in adipose tissue of offspring. FASEB J 2021; 35:e21924. [PMID: 34582059 DOI: 10.1096/fj.202100623r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/11/2021] [Accepted: 08/31/2021] [Indexed: 12/31/2022]
Abstract
Breastmilk miRNAs may act as epigenetic regulators of metabolism and energy homeostasis in offspring. Here, we aimed to investigate the regulatory effects of miR-26a on adipose tissue development. First, the 3T3-L1 cell model was used to identify putative target genes for miR-26a. Then, target genes were analysed in adipose tissue of offspring from dams that supplied lower levels of breastmilk miR-26a to determine whether miR-26a milk concentration might have a long-lasting impact on adipose tissue in the progeny. In the in vitro model, both over- and under-expression of miR-26a were induced by transfecting into 3T3-L1 with miR-26a mimic and inhibitor. Array analysis was performed after induction of miR-26a to ascertain the impact on mRNA target genes and influence of differentiation status. Focusing on genes related to adipose tissue development, transfection with miR-26a mimic reduced the expression of Pten, Hmga1, Stk11, Rb1, and Adam17 in both pre- and mature adipocytes. Data mostly confirmed the results found in the animal model. After weaning, descendants of cafeteria-fed dams breastfed with lower levels of miR-26a displayed greater expression of Hmag1, Rb1, and Adam17 in retroperitoneal white adipose tissue in comparison with controls. Hence, alterations in the amount of miR-26a supplied through milk during lactation is able to alter the expression of target genes in the descendants and may affect adipose tissue development. Thus, milk miR-26a may act as an epigenetic regulator influencing early metabolic program in the progeny, which emerges as a relevant component of an optimal milk composition for correct development.
Collapse
Affiliation(s)
- Catalina A Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, Palma, Spain.,Instituto de Investigación Sanitaria Illes Balears, IdISBa, Palma, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Francisca Serra
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, Palma, Spain.,Instituto de Investigación Sanitaria Illes Balears, IdISBa, Palma, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, Palma, Spain.,Instituto de Investigación Sanitaria Illes Balears, IdISBa, Palma, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Juana Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands, Palma, Spain.,Instituto de Investigación Sanitaria Illes Balears, IdISBa, Palma, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
279
|
Deng YL, Chen S, Wang HT, Wang B, Xiao K. Prescription of Sageretia hamosa Brongn Relieved Goiter through Promoted Apoptosis of Thyroid Cells via miR-511-3p and PTEN/PI3K/Akt Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3506559. [PMID: 34630982 PMCID: PMC8494543 DOI: 10.1155/2021/3506559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/05/2021] [Accepted: 09/08/2021] [Indexed: 02/05/2023]
Abstract
Goiter is thyroid enlargement, in China, Sageretia hamosa Brongn (SHB) can be used to treat goiter, but it has not been reported. Therefore, data analytics of SHB prescription on thyroid were explored in this study to provide a theoretical support for SHB in the treatment of goiter. In this study, rat in goiter model was constructed by using propylthiouracil (PTU) and treated with SHB prescription. Thyroid function about the triiodothyronine (T3), free thyroxine (T4), free triiodothyronine (FT3), free thyroxine (FT4), and thyroid-stimulating hormone (TSH) were measured by ELISA; thyroid coefficient was calculated after weighed thyroid; and HE staining was applied to assess the morphology of thyroid tissue. miRNA microarrays were employed to detect miRNA expression in thyroid tissue of rats. Expression of miR-511-3p was measured by RT-qPCR; expression of proteins (PTEN and apoptosis-related proteins) was tested by western blotting; relationship between miR-511-3p and PTEN was investigated by dual luciferase reporter gene assay; cell viability rate was determined by CCK-8; and cell cycle distribution and apoptosis rate were detected by flow cytometry. The results showed that SHB prescription ameliorated goiter and downregulated miR-511-3p. miR-511-3p targeted PTEN in thyroid cells and PTEN negatively regulated the activation of PI3K/Akt pathway. Furthermore, the inhibition of apoptosis in thyroid cells caused by the overexpression of miR-511-3p or the activation of PI3K/Akt pathway was reversed by treatment of SHB prescription, inhibition of miR-511-3p, or overexpression of PTEN. In conclusion, SHB prescription promoted apoptosis of thyroid through decreased miR-511-3p and regulated PTEN/PI3K/Akt pathway, it might suggest possible medical applications.
Collapse
Affiliation(s)
- Yang-lin Deng
- Precision Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pharmacy, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Su Chen
- Department of Outpatient, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Hong-tu Wang
- Department of Infections Disease, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Bo Wang
- Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kai Xiao
- Precision Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
280
|
Yuan DD, Jia CD, Yan MY, Wang J. Circular RNA hsa_circ_0000730 restrains cell proliferation, migration, and invasion in cervical cancer through miR-942-5p/PTEN axis. Kaohsiung J Med Sci 2021; 37:964-972. [PMID: 34562344 DOI: 10.1002/kjm2.12443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 06/07/2021] [Accepted: 06/24/2021] [Indexed: 12/19/2022] Open
Abstract
Circular RNAs (circRNAs) play prominent roles in regulating the progression of cancers. This study is aimed to decipher the role of hsa_circ_0000730 in cervical cancer (CC).The differentially expressed circRNAs of CC were screened out from the Gene Expression Omnibus database. qRT-PCR was used to detect circ_0000730 expression in CC tissues and cell lines, and the Kaplan-Meier curve was adopted to figure out the relationship between circ_000730 expression and the overall survival time of CC patients. BrdU assay and Tanswell assay were utilized to examine the proliferation, migration, and invasion of CC cells. Western blot was adopted to detect PTEN protein expression. Bioinformatics analysis and dual-luciferase reporter assay were used to examine the target relationship between miR-942-5p and circ_0000730 or PTEN, respectively.Circ_0000730 was among the differentially expressed circRNAs in CC. Circ_0000730 was significantly down-regulated in the cancer tissues of 50 CC patients and CC cell lines. Additionally, underexpression of circ_0000730 was associated with the shorter survival time of CC patients. Gain- and loss-of-function assays highlighted that circ_0000730 significantly inhibited the proliferation, migration, and invasion of CC cells. Mechanistically, miR-942-5p was identified as a downstream target of circ_0000730, and circ_0000730 could positively regulate PTEN expression via repressing miR-942-5p in CC cells.Circ_0000730 inhibits the proliferation, migration, and invasion of CC cells via regulating miR-942-5p/PTEN axis. Circ_0000730 probably acts as a tumor suppressor in CC, and it may be a candidate target for the treatment of CC.
Collapse
Affiliation(s)
- Dan-Dan Yuan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, PR China
| | - Cun-De Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, PR China
| | - Ming-Yu Yan
- Department of Respiratory, The Third Affiliated Hospital of Inner Mongolia Medical College, Baotou, PR China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, PR China
| |
Collapse
|
281
|
Kwon YM, Kim SH, Jung YS, Kwak JH. Synthesis and Biological Evaluation of ( S)-2-(Substituted arylmethyl)-1-oxo-1,2,3,4-tetrahydropyrazino[1,2- a]indole-3-carboxamide Analogs and Their Synergistic Effect against PTEN-Deficient MDA-MB-468 Cells. Pharmaceuticals (Basel) 2021; 14:ph14100974. [PMID: 34681198 PMCID: PMC8537755 DOI: 10.3390/ph14100974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/07/2023] Open
Abstract
A series of twenty-six compounds of furfuryl or benzyl tetrahydropyrazino[1,2-a]indole analogs were synthesized and evaluated for cytotoxic activity against the estrogen receptor (ER)-positive breast cancer cell line (MCF-7) and the epidermal growth factor receptor (EGFR) over-expressed triple-negative breast cancer cell line (MDA-MB-468). Among them, compounds 2b, 2f and 2i showed more potent activity and selectivity against MDA-MB-468 cells than gefitinib, as an EGFR- tyrosine kinase inhibitor. In addition, it was confirmed by means of isobologram analysis of combinational treatment with gefitinib that they have a synergistic effect, especially compounds 2b and 2f, which inhibit Akt T308 phosphorylation. Moreover, it was confirmed that 2-benzyl-1-oxo-1,2,3,4-tetrahydropyrazino[1,2-a]indole-3-carboxamide analogs (2b, 2f, and Ref 2) tend to selectively inhibit PI3Kβ, which is involved in the phosphorylation of Akt.
Collapse
Affiliation(s)
- Ye-Mi Kwon
- College of Pharmacy, Kyungsung University, Busan 48434, Korea;
| | - Sou Hyun Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Korea;
| | - Young-Suk Jung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Korea;
- Correspondence: (Y.-S.J.); (J.-H.K.); Tel.: +82-51-510-2816 (Y.-S.J.); +82-51-663-4889 (J.-H.K.)
| | - Jae-Hwan Kwak
- College of Pharmacy, Kyungsung University, Busan 48434, Korea;
- Correspondence: (Y.-S.J.); (J.-H.K.); Tel.: +82-51-510-2816 (Y.-S.J.); +82-51-663-4889 (J.-H.K.)
| |
Collapse
|
282
|
Jiang Z, Hou Z, Li L, Liu W, Yu Z, Chen S. Exosomal circEPB41L2 serves as a sponge for miR-21-5p and miR-942-5p to suppress colorectal cancer progression by regulating the PTEN/AKT signalling pathway. Eur J Clin Invest 2021; 51:e13581. [PMID: 34022068 DOI: 10.1111/eci.13581] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Exosomes contain many functional RNAs, including circular RNA (circRNA), which are critical for cancer progression. However, the role of exosomal circEPB41L2 in colorectal cancer (CRC) remains unclear. METHODS Exosomes were isolated from plasma and cells. The characteristics of the exosomes were identified using transmission electron microscopy and nanoparticle tracking analysis. The protein levels of exosome markers and PTEN/AKT-related markers were measured using Western blot analysis. The expression of circEPB41L2, microRNA (miR)-21-5p and miR-942-5p was verified by quantitative real-time PCR. The proliferation, apoptosis, migration and invasion of cells were determined using cell counting kit eight assay, colony formation assay, flow cytometry, wound healing assay and transwell assay. Biotin-labelled RNA pull-down assay, dual-luciferase reporter assay and RIP assay were conducted to evaluate the interaction between circEPB41L2 and miR-21-5p or miR-942-5p. The effects of exosomal circEPB41L2 on colorectal cancer tumour growth were confirmed using animal experiments. RESULTS CircEPB41L2 was downregulated in the exosomes from colorectal cancer patients and cells. Overexpressed circEPB41L2 inhibited colorectal cancer cell proliferation, migration, invasion and promoted apoptosis, as well as suppressed the activity of PTEN/AKT signalling pathway. CircEPB41L2 could sponge miR-21-5p or miR-942-5p. MiR-21-5p or miR-942-5p could reverse the inhibition effect of circEPB41L2 on colorectal cancer progression and PTEN/AKT signalling pathway. In addition, we discovered that circEPB41L2 was mainly located at exosomes. Exosomal circEPB41L2 also could restrain colorectal cancer progression and the activity of PTEN/AKT signalling pathway. Animal experiments suggested that exosomal-mediated circEPB41L2 inhibited colorectal cancer tumour growth. CONCLUSION Our data revealed that exosomal circEPB41L2 sponged miR-21-5p and miR-942-5p to repress colorectal cancer progression by regulating the PTEN/AKT signalling pathway.
Collapse
Affiliation(s)
- Zhipeng Jiang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Zehui Hou
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Liang Li
- Department of Gastrointestinal Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Liu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Zhuomin Yu
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| | - Shuang Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, China
| |
Collapse
|
283
|
Abbas A, Padmanabhan R, Eng C. Metabolic stress regulates genome-wide transcription in a PTEN-dependent manner. Hum Mol Genet 2021; 29:2736-2745. [PMID: 32744308 DOI: 10.1093/hmg/ddaa168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/19/2020] [Accepted: 07/27/2020] [Indexed: 12/31/2022] Open
Abstract
PTEN is implicated in a wide variety of pathophysiological conditions and traditionally studied in the context of the PIK3-AKT-mTOR axis. Recent studies from our group and others have reported a novel role of PTEN in the regulation of transcription at the genome-wide scale. This emerging role of PTEN on global transcriptional regulation is providing a better understanding of various diseases, including cancer. Because cancer progression is an energy-demanding process and PTEN is known to regulate metabolic processes, we sought to understand the role of PTEN in transcriptional regulation under metabolic stress, a condition often developing in the tumor microenvironment. In the present study, we demonstrate that PTEN modulates genome-wide RNA Polymerase II occupancy in cells undergoing glucose deprivation. The glucose-deprived PTEN null cells were found to continue global gene transcription, which may activate a survival mode. However, cells with constitutive PTEN expression slow transcription, an evolutionary mechanism that may save cellular energy and activate programmed cell death pathways, in the absence of glucose. Interestingly, alternative exon usage by PTEN null cells is increased under metabolic stress in contrast to PTEN-expressing cells. Overall, our study demonstrates distinct mechanisms involved in PTEN-dependent genome-wide transcriptional control under metabolic stress. Our findings provide a new insight in understanding tumor pathology and how PTEN loss of function, whether by genetic or non-genetic mechanisms, can contribute to a favorable transcriptional program employed by tumor cells to escape apoptosis, hence developing more aggressive and metastatic phenotypes.
Collapse
Affiliation(s)
- Ata Abbas
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44116, USA
| | - Roshan Padmanabhan
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences.,Germline High Risk Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44116, USA
| |
Collapse
|
284
|
He Y, Jiang S, Mao C, Zheng H, Cao B, Zhang Z, Zhao J, Zeng Y, Mao X. The deubiquitinase USP10 restores PTEN activity and inhibits non-small cell lung cancer cell proliferation. J Biol Chem 2021; 297:101088. [PMID: 34416231 PMCID: PMC8429974 DOI: 10.1016/j.jbc.2021.101088] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) protein is a key player in tumorigenesis of non–small cell lung cancer (NSCLC) and was recently found to be inactivated by tripartite motif containing 25 (TRIM25)–mediated K63-linked polyubiquitination. However, the deubiquitinase (Dub) coordinate TRIM25 in PTEN ubiquitination is still elusive. In the present study, we found that this K63-linked polyubiquitination could be ablated by the ubiquitin-specific protease 10 (USP10) in a screen against a panel of Dubs. We found using coimmununoprecipitation/immunoblotting that USP10 interacted with PTEN and reduced the K63-linked polyubiquitination of PTEN mediated by TRIM25 in NSCLC cells. Moreover, USP10, but not its inactive C424A deubiquitinating mutant or other Dubs, abolished PTEN from K63-linked polyubiquitination mediated by TRIM25. In contrast to TRIM25, USP10 restored PTEN phosphatase activity and reduced the production of the secondary messenger phosphatidylinositol-3,4,5-trisphosphate, thereby inhibiting AKT/mammalian target of rapamycin progrowth signaling transduction in NSCLC cells. Moreover, USP10 was downregulated in NSCLC cell lines and primary tissues, whereas TRIM25 was upregulated. Consistent with its molecular activity, re-expression of USP10 suppressed NSCLC cell proliferation and migration, whereas knockout of USP10 promoted NSCLC cell proliferation and migration. In conclusion, the present study demonstrates that USP10 coordinates TRIM25 to modulate PTEN activity. Specifically, USP10 activates PTEN by preventing its K63-linked polyubiquitination mediated by TRIM25 and suppresses the AKT/mammalian target of rapamycin signaling pathway, thereby inhibiting NSCLC proliferation, indicating that it may be a potential drug target for cancer treatment.
Collapse
Affiliation(s)
- Yuanming He
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shuoyi Jiang
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Chenyu Mao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hui Zheng
- Institute of Biomedical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Biyin Cao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zubin Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuanying Zeng
- Department of Oncology, Suzhou Municipal Hospital, Suzhou, China.
| | - Xinliang Mao
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Diseases, The Second Affiliated Hospital, Guangdong Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
285
|
Ming Y, Liu ZP. Overexpression of lncRNA-NEF regulates the miR-155/PTEN axis to inhibit adipogenesis and promote osteogenesis. Kaohsiung J Med Sci 2021; 37:930-939. [PMID: 34382731 DOI: 10.1002/kjm2.12423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/11/2021] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Osteoporosis is characterized by osteopenia and bone tissue microstructure degradation. Adipose-derived stem cells (ADSCs) are multipotent adult stem cells that have the ability to yield mesenchymal stem cells and have the potential to undergo osteogenesis and bone regeneration. Therefore, ADSCs have the potential to treat osteoporosis, but the molecular mechanism of these cells in the process of osteogenesis and osteoclasts is still not clear. In the present study, we collected serum samples from 10 clinical osteoporosis patients to detect long noncoding RNA-neighboring enhancer of FOXA2 (lncRNA-NEF) and miR-155 expression levels. Half of these patients were senile and half were postmenopausal women, and nine of them have used steroids for a long time, in which ADSCs were cultured and induced to adipogenic and osteogenic differentiations. Quantitative real-time polymerase chain reaction was used to detect the expression of genes in ADSCs. Overexpression of lncRNA-NEF in ADSCs were undertaken to verify its regulatory function on cell osteogenic and adipogenic differentiations. A luciferase activity experiment was performed to determine the relationship between miR-155 and phosphatase and tensin homologue deleted on chromosome 10 (PTEN). The level of lncRNA-NEF was downregulated, and miR-155 was upregulated, in serum samples from patients with clinical osteoporosis. LncRNA-NEF showed different expression levels in the induction of osteogenic or adipogenic differentiation, which increased during osteogenic induction and decreased during adipogenic induction. Overexpression of lncRNA-NEF or downregulation of miR-155 in ADSCs promoted osteogenic differentiation and inhibited adipogenesis progression. PTEN was the direct target of miR-155 and was involved in the regulation of osteogenic differentiation. Overexpression of lncRNA-NEF regulated the miR-155/PTEN axis to inhibit adipogenesis and promote osteogenesis in ADSCs.
Collapse
Affiliation(s)
- Ying Ming
- Department of Clinical Laboratory, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, China
| | - Zheng-Peng Liu
- Department of Spinal Surgery Ward, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, China
| |
Collapse
|
286
|
Ghaznavi H, Shirvaliloo M, Zarebkohan A, Shams Z, Radnia F, Bahmanpour Z, Sargazi S, Saravani R, Shirvalilou S, Shahraki O, Shahraki S, Nazarlou Z, Sheervalilou R. An Updated Review on Implications of Autophagy and Apoptosis in Tumorigenesis: Possible Alterations in Autophagy through Engineered Nanomaterials and Their Importance in Cancer Therapy. Mol Pharmacol 2021; 100:119-143. [PMID: 33990406 DOI: 10.1124/molpharm.121.000234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Most commonly recognized as a catabolic pathway, autophagy is a perplexing mechanism through which a living cell can free itself of excess cytoplasmic components, i.e., organelles, by means of certain membranous vesicles or lysosomes filled with degrading enzymes. Upon exposure to external insult or internal stimuli, the cell might opt to activate such a pathway, through which it can gain control over the maintenance of intracellular components and thus sustain homeostasis by intercepting the formation of unnecessary structures or eliminating the already present dysfunctional or inutile organelles. Despite such appropriateness, autophagy might also be considered a frailty for the cell, as it has been said to have a rather complicated role in tumorigenesis. A merit in the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. In fact, several investigations on tumorigenesis have reported diminished levels of autophagic activity in tumor cells, which might result in transition to malignancy. On the contrary, autophagy has been suggested to be a seemingly favorable mechanism to progressed malignancies, as it contributes to survival of such cells. Based on the recent literature, this mechanism might also be activated upon the entry of engineered nanomaterials inside a cell, supposedly protecting the host from foreign materials. Accordingly, there is a good chance that therapeutic interventions for modulating autophagy in malignant cells using nanoparticles may sensitize cancerous cells to certain treatment modalities, e.g., radiotherapy. In this review, we will discuss the signaling pathways involved in autophagy and the significance of the mechanism itself in apoptosis and tumorigenesis while shedding light on possible alterations in autophagy through engineered nanomaterials and their potential therapeutic applications in cancer. SIGNIFICANCE STATEMENT: Autophagy has been said to have a complicated role in tumorigenesis. In the early stages of tumor formation, autophagy appears to be salutary because of its tumor-suppressing effects. On the contrary, autophagy has been suggested to be a favorable mechanism to progressed malignancies. This mechanism might be affected upon the entry of nanomaterials inside a cell. Accordingly, therapeutic interventions for modulating autophagy using nanoparticles may sensitize cancerous cells to certain therapies.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Milad Shirvaliloo
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Amir Zarebkohan
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zinat Shams
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Fatemeh Radnia
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Zahra Bahmanpour
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Saman Sargazi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ramin Saravani
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sakine Shirvalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Sheida Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Ziba Nazarlou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (H.G.), Infectious and Tropical Diseases Research Center, (M.S.), Department of Medical Nanotechnology, School of Advanced Medical Sciences,Tabriz University of Medical Sciences, Tabriz, Iran (A.Z.), Department of Biological Science, Kharazmi University, Tehran, Iran (Z.S.), Department of Medical Biotechnology, Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran (F.R.), Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran (Z.B.), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sar), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (R.S.), Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran (S.Sh), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (O.S), Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (S.Sha), Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey (Z.N.), Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran (R.Sh)
| |
Collapse
|
287
|
Montella L, Sarno F, Altucci L, Cioffi V, Sigona L, Di Colandrea S, De Simone S, Marinelli A, Facchini BA, De Vita F, Berretta M, de Falco R, Facchini G. A Root in Synapsis and the Other One in the Gut Microbiome-Brain Axis: Are the Two Poles of Ketogenic Diet Enough to Challenge Glioblastoma? Front Nutr 2021; 8:703392. [PMID: 34422883 PMCID: PMC8378133 DOI: 10.3389/fnut.2021.703392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/29/2021] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma is the most frequent and aggressive brain cancer in adults. While precision medicine in oncology has produced remarkable progress in several malignancies, treatment of glioblastoma has still limited available options and a dismal prognosis. After first-line treatment with surgery followed by radiochemotherapy based on the 2005 STUPP trial, no significant therapeutic advancements have been registered. While waiting that genomic characterization moves from a prognostic/predictive value into therapeutic applications, practical and easy-to-use approaches are eagerly awaited. Medical reports on the role of the ketogenic diet in adult neurological disorders and in glioblastoma suggest that nutritional interventions may condition outcomes and be associated with standard therapies. The acceptable macronutrient distribution of daily calories in a regular diet are 45-65% of daily calories from carbohydrates, 20-35% from fats, and 10-35% from protein. Basically, the ketogenic diet follows an approach based on low carbohydrates/high fat intake. In carbohydrates starvation, body energy derives from fat storage which is used to produce ketones and act as glucose surrogates. The ketogenic diet has several effects: metabolic interference with glucose and insulin and IGF-1 pathways, influence on neurotransmission, reduction of oxidative stress and inflammation, direct effect on gene expression through epigenetic mechanisms. Apart from these central effects working at the synapsis level, recent evidence also suggests a role for microbiome and gut-brain axis induced by a ketogenic diet. This review focuses on rationales supporting the ketogenic diet and clinical studies will be reported, looking at future possible perspectives.
Collapse
Affiliation(s)
- Liliana Montella
- Medical Oncology Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Federica Sarno
- Precision Medicine Department, “Luigi Vanvitelli” University of Campania, Naples, Italy
| | - Lucia Altucci
- Precision Medicine Department, “Luigi Vanvitelli” University of Campania, Naples, Italy
| | - Valentina Cioffi
- Neurosurgery Operative Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Luigi Sigona
- Neurosurgery Operative Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Salvatore Di Colandrea
- Department of Emergency and Critical Care, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Stefano De Simone
- Medical Oncology Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Alfredo Marinelli
- Operative Unit Neuroncology University Federico II, Naples, Italy
- Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Neuromed Istituto Neurologico Mediterraneo (INM), Isernia, Italy
| | - Bianca Arianna Facchini
- Division of Medical Oncology, Precision Medicine Department, “Luigi Vanvitelli” University of Campania, Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Precision Medicine Department, “Luigi Vanvitelli” University of Campania, Naples, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Raffaele de Falco
- Neurosurgery Operative Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| | - Gaetano Facchini
- Medical Oncology Complex Unit, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, Naples, Italy
| |
Collapse
|
288
|
Increased effects of 2,5-dimethylcelecoxib on sensitivity of hepatocellular carcinoma cells to sorafenib via CYP3A5 expression and activation of AMPK. Toxicol In Vitro 2021; 76:105226. [PMID: 34293431 DOI: 10.1016/j.tiv.2021.105226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 11/21/2022]
Abstract
As the occurrence and development of HCC are often accompanied by inflammation, the combination of sorafenib with other therapeutic drugs, especially anti-inflammatory drugs, is one of the directions to be explored at present. Our previous research has been focused on the anti-inflammatory drug 2,5-dimethylcelecoxib (DMC), whether DMC combined with sorafenib could elevate the effect of inhibiting HCC deserves further exploration. In this study, we found that DMC induced CYP3A5 expression in HCC cells in a time-dependent and concentration dependent manner. We observed that sorafenib inhibited CYP3A5 expression in liver cancer cells, and activated the phosphorylation of Akt. Upregulated CYP3A5 and DMC treatment enhanced the ability of sorafenib to inhibit migration. The combination of DMC with sorafenib had a synergistic effect of enhancing drug sensitivity (CI < 1), meanwhile, inhibited the proliferation and promoted apoptosis of HCC. Activation of the AMPK pathway and inhibition of the PI3K/Akt pathway were observed in cells treated with DMC in combination with sorafenib and could be reverted by an AMPK pathway inhibitor. Our findings suggest that DMC induces CYP3A5 expression and enhances the anticancer effect of sorafenib by activating AMPK, which would be a novel strategy for drug combination to prevent drug resistance.
Collapse
|
289
|
Liu S, Ma H, Zhang H, Deng C, Xin P. Recent advances on signaling pathways and their inhibitors in rheumatoid arthritis. Clin Immunol 2021; 230:108793. [PMID: 34242749 DOI: 10.1016/j.clim.2021.108793] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by systemic synovitis leading to joint destruction in which imbalances in pro-inflammatory and anti-inflammatory cytokines promote the induction of autoimmunity. Some pro-inflammatory cytokines can trigger the signaling pathways which responsible for immune-mediated inflammation in RA, and the activated signaling pathways produce pro-inflammatory cytokines, resulting in aggravation of RA. Hence, understanding of the signaling pathways and their inhibitors might be advantageous in the development of therapeutic targets and new drugs for RA. In the current review, we summarize the signaling pathways involved in the pathogenesis of RA as well as the potential role of specific inhibitors in its management. We hope this paper may serve a reference for future studies on signaling pathways implicated in the pathogenesis of RA and benefit the treatment of RA.
Collapse
Affiliation(s)
- Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, China
| | - Huaxi Zhang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
290
|
Zhan H, Bai Y, Lv Y, Zhang X, Zhang L, Deng S. Pharmacological mechanism of mylabris in the treatment of leukemia based on bioinformatics and systematic pharmacology. Bioengineered 2021; 12:3229-3239. [PMID: 34224300 PMCID: PMC8806889 DOI: 10.1080/21655979.2021.1943110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Leukemia is a common blood cancer, whose treatment usually necessitates chemo/radiotherapy and bone marrow transplant. Hence, safer and more effective options are urgently needed. Mylabris, the dried body of blister beetles, has been used extensively in traditional Chinese medicine. This study applied bioinformatics and systematic pharmacology to investigate the mechanism of action of mylabris in the treatment of leukemia. Five effective components and 35 corresponding target proteins were identified by screening the TCMSP database; whereas 776 genes related to leukemia were selected using OMIM, GeneCards, and the Therapeutic Target Database. Eight genes common to mylabris and leukemia were identified. Protein-protein interaction network analysis and a component-target-pathway diagram identified TP53 and PTEN as key gene targets of mylabris in the treatment of leukemia. GO enrichment analysis pointed to DNA damage and cell cycle disorder caused by p53 signaling as the most significant processes; whereas KEGG enrichment pointed to the p53 signaling pathway. In summary, mylabris may exert a therapeutic effect on leukemia by triggering DNA damage, inducing apoptosis, as well as inhibiting the growth and proliferation of tumor cells through the regulation of TP53 and PTEN. These findings provide a mechanistic rationale for the treatment of leukemia with traditional Chinese medicine.
Collapse
Affiliation(s)
- Huali Zhan
- Department of Humanities and Social Sciences, Zhejiang Industry Polytechnic College, Zhejiang, China
| | - Yujiao Bai
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China.,School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yu Lv
- Wenzhou Medical University Renji College, Wenzhou, Zhejiang, China
| | - Xianqin Zhang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Lin Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Pharmacy, Shaoxing People's Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang, China
| | - Shanshan Deng
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China.,School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
291
|
The role of Hypoxia-Inducible Factor-1alpha and its signaling in melanoma. Biomed Pharmacother 2021; 141:111873. [PMID: 34225012 DOI: 10.1016/j.biopha.2021.111873] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Adaptation to the loss of O2 is regulated via the activity of hypoxia-inducible factors such as Hypoxia-Inducible Factor-1 (HIF-1). HIF-1 acts as a main transcriptional mediator in the tissue hypoxia response that regulates over 1000 genes related to low oxygen tension. The role of HIF-1α in oncogenic processes includes angiogenesis, tumor metabolism, cell proliferation, and metastasis, which has been examined in various malignancies, such as melanoma. Melanoma is accompanied by a high death rate and a cancer type whose incidence has risen over the last decades. The linkage between O2 loss and melanogenesis had extensively studied over decades. Recent studies revealed that HIF-1α contributes to melanoma progression via different signaling pathways such as PI3K/Akt/mTOR, RAS/RAF/MEK/ERK, JAK/STAT, Wnt/β-catenin, Notch, and NF-κB. Also, various microRNAs (miRs) are known to mediate the HIF-1α role in melanoma. Therefore, HIF-1α offers a diagnostic/prognostic biomarker and a candidate for targeted therapy in melanoma.
Collapse
|
292
|
Alkafaji HA, Raji A, Rahman HS, Zekiy AO, Adili A, Jalili M, Hojjatipour T, Cid‐Arregui A, Shomali N, Tarzi S, Tamjidifar R, Heshmati R, Marofi F, Akbari M, Hasanzadeh A, Deljavanghodrati M, Jarahian M, Sandoghchian Shotorbani S. Up-regulation of KISS1 as a novel target of Let-7i in melanoma serves as a potential suppressor of migration and proliferation in vitro. J Cell Mol Med 2021; 25:6864-6873. [PMID: 34096173 PMCID: PMC8278109 DOI: 10.1111/jcmm.16695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma is a kind of skin cancer that is begun by the alteration of melanocytes. miRNAs are small non-coding RNA molecules that regulate a variety of biological processes. KISS1, the metastasis-suppressor gene, encodes kisspeptins which inhibits migration and proliferation of cancers. This study was aimed to determine the role of Let-7i and KISS1 in melanoma cell migration and proliferation. At first, the expression of Let-7i and KISS1 was determined in patients with melanoma. In the in vitro part of the study, Let-7i mimics were transfected and the impact of its restoration on target gene expression, proliferation, migration and apoptosis of SK-MEL-3 melanoma cell line was assessed by real-time PCR and Western blotting, MTT assay, wound-healing assay and flow cytometry, respectively. Besides, KISS1 inhibitor siRNA alone and along with Let-7i was transfected to determine their probable correlation. The results revealed that either Let-7i or KISS1 were down-regulated in patients with melanoma. The results obtained from the in vitro part of the study revealed that restoration of Let-7i reduced the expression of metastasis- and proliferation-related target genes. Moreover, it was revealed that up-regulation of Let-7i attenuated migration and proliferation capability of SK-MEL-3 cells. Besides, it was demonstrated that Let-7i restoration induced apoptosis in melanoma cells. More importantly, the KISS1 inhibitor caused a prominent cell migration and proliferation, attenuated by Let-7i re-expression. To sum up, the present study revealed the impressive role of Let-7i restoration along with its correlation with KISS1 on melanoma carcinogenicity which may be applicable in future in vivo studies.
Collapse
Affiliation(s)
| | - Ahmed Raji
- College of medicineUniversity of BabylonBabylonIraq
| | - Heshu S. Rahman
- Department of PhysiologyCollege of MedicineUniversity of SuleimanyahSuleimanyahIraq
| | - Angelina O. Zekiy
- Sechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Ali Adili
- Department of OncologyTabriz University of Medical SciencesTabrizIran
| | | | - Tahereh Hojjatipour
- Department of Hematology and Blood TransfusionStudents Research CentreSchool of Allied MedicineTehran University of Medical SciencesTehranIran
| | - Angel Cid‐Arregui
- Targeted Tumor Vaccines UnitGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Navid Shomali
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Saeed Tarzi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Rozita Tamjidifar
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Ramin Heshmati
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Faroogh Marofi
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Morteza Akbari
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Ali Hasanzadeh
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | | | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401)German Cancer Research CenterHeidelbergGermany
| | - Siamak Sandoghchian Shotorbani
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
293
|
Banfi F, Rubio A, Zaghi M, Massimino L, Fagnocchi G, Bellini E, Luoni M, Cancellieri C, Bagliani A, Di Resta C, Maffezzini C, Ianielli A, Ferrari M, Piazza R, Mologni L, Broccoli V, Sessa A. SETBP1 accumulation induces P53 inhibition and genotoxic stress in neural progenitors underlying neurodegeneration in Schinzel-Giedion syndrome. Nat Commun 2021; 12:4050. [PMID: 34193871 PMCID: PMC8245514 DOI: 10.1038/s41467-021-24391-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.
Collapse
MESH Headings
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/metabolism
- Abnormalities, Multiple/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Craniofacial Abnormalities/genetics
- Craniofacial Abnormalities/metabolism
- Craniofacial Abnormalities/pathology
- DNA Damage
- Hand Deformities, Congenital/genetics
- Hand Deformities, Congenital/metabolism
- Hand Deformities, Congenital/pathology
- Heredodegenerative Disorders, Nervous System/genetics
- Heredodegenerative Disorders, Nervous System/metabolism
- Heredodegenerative Disorders, Nervous System/pathology
- Humans
- Intellectual Disability/genetics
- Intellectual Disability/metabolism
- Intellectual Disability/pathology
- Mutation
- Nails, Malformed/genetics
- Nails, Malformed/metabolism
- Nails, Malformed/pathology
- Neural Stem Cells/metabolism
- Neural Stem Cells/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organoids
- Tumor Suppressor Protein p53/antagonists & inhibitors
Collapse
Affiliation(s)
- Federica Banfi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Alicia Rubio
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Fagnocchi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Edoardo Bellini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mirko Luoni
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Cancellieri
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Human Induced Pluripotent Stem Cells service, Istituto Italiano di Oncologia Molecolare (IFOM), Milan, Italy
| | - Anna Bagliani
- Medical Oncology Unit, ASST Ovest Milanese, Legnano Hospital, Legnano, Italy
| | - Chiara Di Resta
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Genomics for human disease diagnosis, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Maffezzini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Ianielli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | | | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
294
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
295
|
Zhang S, Xu J, Chen Q, Zhang F, Wang H, Guo H. lncRNA MT1JP-overexpression abolishes the silencing of PTEN by miR-32 in hepatocellular carcinoma. Oncol Lett 2021; 22:604. [PMID: 34188706 PMCID: PMC8227557 DOI: 10.3892/ol.2021.12865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/27/2021] [Indexed: 01/17/2023] Open
Abstract
Previous studies have shown that long non-coding RNA (lncRNA) MT1JP plays a role as a tumor suppressor in several types of cancer. The present study aimed to explore the role of MT1JP in hepatocellular carcinoma (HCC). Paired HCC and non-tumor tissues from 64 patients with HCC were subjected to RNA isolation and reverse transcription-quantitative PCR (RT-qPCR) to analyze the differential expression of MT1JP, microRNA (miR)-32 and phosphatase and tensin homolog (PTEN) in HCC. Cell transfections, followed by RT-qPCR and western blotting, were carried out to investigate the interactions among MT1JP, miR-32 and PTEN. The role of MT1JP, miR-32 and PTEN in regulating HCC cell proliferation was assessed using a Cell Counting Kit-8 assay. It was found that MT1JP was downregulated in HCC cancer tissues compared with that in non-cancer tissues. Survival analysis showed that patients with low MT1JP expression levels exhibited a significantly higher 5-year overall survival rate compared with patients with high MT1JP levels. The expression of MT1JP in HCC tissues was positively associated with PTEN and negatively associated with miR-32. Overexpression of MT1JP increased the expression levels of PTEN and decreased the expression levels of miR-32. Overexpression of miR-32 did not affect the expression of MT1JP but decreased the expression levels of PTEN and attenuated the effect of overexpression of MT1JP on the expression of PTEN. Overexpression of MT1JP and PTEN decreased the proliferation of HCC cells. Overexpression of miR-32 played an opposite role and attenuated the effects of overexpression of MT1JP. Therefore, MT1JP may upregulate PTEN by downregulating miR-32 to regulate HCC cell proliferation.
Collapse
Affiliation(s)
- Shuhua Zhang
- Department of Hepatobiliary Surgery, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jianqun Xu
- Department of Respiratory Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Chen
- Department of Hepatobiliary Surgery, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fan Zhang
- Department of Hepatobiliary Surgery, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongjuan Wang
- Department of Respiratory Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongrong Guo
- Department of Respiratory Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
296
|
Rao D, Yu C, Sheng J, Lv E, Huang W. The Emerging Roles of circFOXO3 in Cancer. Front Cell Dev Biol 2021; 9:659417. [PMID: 34150756 PMCID: PMC8213346 DOI: 10.3389/fcell.2021.659417] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
Circular RNAs (circRNAs) are a class of endogenous non-coding RNAs which are mainly formed by reverse splicing of precursor mRNAs. They are relatively stable and resistant to RNase R because of their covalently closed structure without 5' caps or 3' poly-adenylated tails. CircRNAs are widely expressed in eukaryotic cells and show tissue, timing, and disease specificity. Recent studies have found that circRNAs play an important role in many diseases. In particular, they affect the proliferation, invasion and prognosis of cancer by regulating gene expression. CircRNA Forkhead box O3 (circFOXO3) is a circRNA confirmed to be abnormally expressed in a variety of cancers, including prostate cancer, hepatocellular carcinoma, glioblastoma, bladder cancer, and breast cancer, etc. At present, the feature of circFOXO3 as a molecular sponge is widely studied to promote or inhibit the development of cancers. However, the diverse functions of circFOXO3 have not been fully understood. Hence, it is important to review the roles of circFOXO3 in cancers. This review has summarized and discussed the roles and molecular mechanism of circFOXO3 and its target genes in these cancers, which can help to enrich our understanding to the functions of circRNAs and carry out subsequent researches on circFOXO3.
Collapse
Affiliation(s)
- Dean Rao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengpeng Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Sheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Enjun Lv
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
297
|
Competing Endogenous RNAs in Cervical Carcinogenesis: A New Layer of Complexity. Processes (Basel) 2021. [DOI: 10.3390/pr9060991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression by binding to complementary sequences within target mRNAs. Apart from working ‘solo’, miRNAs may interact in important molecular networks such as competing endogenous RNA (ceRNA) axes. By competing for a limited pool of miRNAs, transcripts such as long noncoding RNAs (lncRNAs) and mRNAs can regulate each other, fine-tuning gene expression. Several ceRNA networks led by different lncRNAs—described here as lncRNA-mediated ceRNAs—seem to play essential roles in cervical cancer (CC). By conducting an extensive search, we summarized networks involved in CC, highlighting the major impacts of such dynamic molecular changes over multiple cellular processes. Through the sponging of distinct miRNAs, some lncRNAs as HOTAIR, MALAT1, NEAT1, OIP5-AS1, and XIST trigger crucial molecular changes, ultimately increasing cell proliferation, migration, invasion, and inhibiting apoptosis. Likewise, several lncRNAs seem to be a sponge for important tumor-suppressive miRNAs (as miR-140-5p, miR-143-3p, miR-148a-3p, and miR-206), impairing such molecules from exerting a negative post-transcriptional regulation over target mRNAs. Curiously, some of the involved mRNAs code for important proteins such as PTEN, ROCK1, and MAPK1, known to modulate cell growth, proliferation, apoptosis, and adhesion in CC. Overall, we highlight important lncRNA-mediated functional interactions occurring in cervical cells and their closely related impact on cervical carcinogenesis.
Collapse
|
298
|
Zhu L, Chen R, Jiang C, Xie Q, Zhao W, Gao X, Huang H. Mechanism underlying long non‑coding RNA ILF3‑AS1‑mediated inhibition of cervical cancer cell proliferation, invasion and migration, and promotion of apoptosis. Mol Med Rep 2021; 24:554. [PMID: 34080029 PMCID: PMC8188751 DOI: 10.3892/mmr.2021.12193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/24/2020] [Indexed: 01/13/2023] Open
Abstract
Long non-coding RNA ILF3 divergent transcript (ILF3-AS1) displays a tumor-suppressing effect. StarBase predicted that the potential target microRNA (miR) of ILF3-AS1 was miR-454-3p; therefore, the present study investigated the effect of ILF3-AS1 and its target miR-454-3p on cervical cancer (CC). Gene Expression Profiling Interactive Analysis was used to predict the expression of ILF3-AS1 in CC and the overall survival rate of patients. The present study demonstrated that ILF3-AS1 expression was significantly downregulated in human CC tissues and cells compared with adjacent tissues (ANTs) and normal cervical epithelial cells (NCEs), respectively. Patients with CC with high ILF3-AS1 expression displayed higher survival rates compared with patients with low ILF3-AS1 expression. Cell viability, apoptosis, migration and invasion were detected by performing Cell Counting Kit-8, flow cytometry, wound healing and Transwell assays, respectively. Compared with the negative control (NC) group, ILF3-AS1 overexpression significantly inhibited CC cell viability and migration, but significantly increased CC cell apoptosis. Moreover, ILF3-AS1 overexpression significantly upregulated E-Cadherin expression levels, but significantly downregulated N-Cadherin and snail family transcriptional repressor 1 expression levels compared with the NC group. miR-454-3p expression was negatively correlated with ILF3-AS1, and highly expressed in CC tissues and cells compared with ANTs and NCEs, respectively. PTEN, which was predicted and verified as the target gene for miR-454-3p, was significantly downregulated in CC tissues and cells compared with ANTs and NCEs, respectively. ILF3-AS1 expression was positively correlated with PTEN expression, and ILF3-AS1 overexpression partially reversed the inhibitory effect of miR-454-3p on PTEN expression. In conclusion, the present study indicated that ILF3-AS1 inhibited CC cell proliferation and migration, and promoted CC cell apoptosis by inhibiting epithelial-mesenchymal transition, and ILF3-AS1 overexpression partially reversed the inhibitory effect of miR-454-3p on PTEN expression.
Collapse
Affiliation(s)
- Linmei Zhu
- Department of Obstetrics and Gynecology, Guangdong Clifford Hospital, Guangzhou, Guangdong 511495, P.R. China
| | - Ruixia Chen
- Department of Anesthesiology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chunlin Jiang
- Department of Obstetrics and Gynecology, Guangdong Clifford Hospital, Guangzhou, Guangdong 511495, P.R. China
| | - Qingsheng Xie
- Department of Gynecology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Wenshuai Zhao
- Department of Anesthesiology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaohong Gao
- Department of Obstetrics and Gynecology, Guangdong Clifford Hospital, Guangzhou, Guangdong 511495, P.R. China
| | - Haiming Huang
- Department of Anesthesiology, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
299
|
Hussein NA, Malla S, Pasternak MA, Terrero D, Brown NG, Ashby CR, Assaraf YG, Chen ZS, Tiwari AK. The role of endolysosomal trafficking in anticancer drug resistance. Drug Resist Updat 2021; 57:100769. [PMID: 34217999 DOI: 10.1016/j.drup.2021.100769] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/10/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
Multidrug resistance (MDR) remains a major obstacle towards curative treatment of cancer. Despite considerable progress in delineating the basis of intrinsic and acquired MDR, the underlying molecular mechanisms remain to be elucidated. Emerging evidences suggest that dysregulation in endolysosomal compartments is involved in mediating MDR through multiple mechanisms, such as alterations in endosomes, lysosomes and autophagosomes, that traffic and biodegrade the molecular cargo through macropinocytosis, autophagy and endocytosis. For example, altered lysosomal pH, in combination with transcription factor EB (TFEB)-mediated lysosomal biogenesis, increases the sequestration of hydrophobic anti-cancer drugs that are weak bases, thereby producing an insufficient and off-target accumulation of anti-cancer drugs in MDR cancer cells. Thus, the use of well-tolerated, alkalinizing compounds that selectively block Vacuolar H⁺-ATPase (V-ATPase) may be an important strategy to overcome MDR in cancer cells and increase chemotherapeutic efficacy. Other mechanisms of endolysosomal-mediated drug resistance include increases in the expression of lysosomal proteases and cathepsins that are involved in mediating carcinogenesis and chemoresistance. Therefore, blocking the trafficking and maturation of lysosomal proteases or direct inhibition of cathepsin activity in the cytosol may represent novel therapeutic modalities to overcome MDR. Furthermore, endolysosomal compartments involved in catabolic pathways, such as macropinocytosis and autophagy, are also shown to be involved in the development of MDR. Here, we review the role of endolysosomal trafficking in MDR development and discuss how targeting endolysosomal pathways could emerge as a new therapeutic strategy to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Noor A Hussein
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Mariah A Pasternak
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Noah G Brown
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA.
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA; Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, 43614, OH, USA.
| |
Collapse
|
300
|
Wu Y, Ma Y, Li J, Zhou XL, Li L, Xu PX, Li XR, Xue M. The bioinformatics and metabolomics research on anti-hypoxic molecular mechanisms of Salidroside via regulating the PTEN mediated PI3K/Akt/NF-κB signaling pathway. Chin J Nat Med 2021; 19:442-453. [PMID: 34092295 DOI: 10.1016/s1875-5364(21)60043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Indexed: 12/08/2022]
Abstract
Salidroside (SAL), a major bioactive compound of Rhodiola crenulata, has significant anti-hypoxia effect, however, its underlying molecular mechanism has not been elucidated. In order to explore the protective mechanism of SAL, the lactate dehydrogenase (LDH), reactive oxygen species (ROS), superoxide dismutase (SOD) and hypoxia-induced factor 1α (HIF-1α) were measured to establish the PC12 cell hypoxic model. Cell staining and cell viability analyses were performed to evaluate the protective effects of SAL. The metabolomics and bioinformatics methods were used to explore the protective effects of salidroside under hypoxia condition. The metabolite-protein interaction networks were further established and the protein expression level was examined by Western blotting. The results showed that 59 endogenous metabolites changed and the expression of the hub proteins of CK2, p-PTEN/PTEN, PI3K, p-Akt/Akt, NF-κB p65 and Bcl-2 were increased, suggesting that SAL could increase the expression of CK2, which induced the phosphorylation and inactivation of PTEN, reduced the inhibitory effect on PI3K signaling pathways and activated the PI3K/Akt/NF-κB survival signaling pathway. Our study provided an important insight to reveal the protective molecular mechanism of SAL as a novel drug candidate.
Collapse
Affiliation(s)
- Yi Wu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yi Ma
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing Li
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xue-Lin Zhou
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Nerve System Drugs, Beijing 100053, China
| | - Lei Li
- Central Laboratory, Capital Medical University, Beijing 100069, China
| | - Ping-Xiang Xu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Nerve System Drugs, Beijing 100053, China
| | - Xiao-Rong Li
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Nerve System Drugs, Beijing 100053, China.
| | - Ming Xue
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Beijing Engineering Research Center for Nerve System Drugs, Beijing 100053, China.
| |
Collapse
|