1
|
Rodríguez-García A, Ancos-Pintado R, García-Vicente R, Ortiz-Ruiz A, Arroyo A, Navarro MÁ, Morales ML, Guevara-Ramirez P, Justo P, López-Muñoz N, Sánchez-Pina J, Alonso R, Selma MV, Frutos-Lisón MD, García-Villalba R, Tomás-Barberán FA, Ayala R, Martínez-López J, Linares M. Microbiota-derived urolithin A in monoclonal gammopathies and multiple myeloma therapy. MICROBIOME 2025; 13:56. [PMID: 40022244 PMCID: PMC11869585 DOI: 10.1186/s40168-025-02045-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/21/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Gut microbiota-derived urolithins may influence multiple myeloma (MM) disease progression and treatment. We analyzed urolithins and their associated microbiota in a retrospective cohort of 45 patients with active MM or premalignant disease using mass spectrometry and 16S rRNA gene sequencing. RESULTS Patients with detectable levels of urolithin in serum and stool and a higher abundance of urolithin-related microbiota had a better outcome. Analysis of the effects of urolithin A (UroA) treatment ex vivo, in vitro, and in vivo revealed that UroA is cytotoxic against MM cell lines and modulates the cell cycle and mitochondrial activity. Notably, UroA inhibits the proliferation of primary MM cells in vitro and in a xenograft mouse model, improving overall survival. Finally, combination therapy with UroA and bortezomib has a synergistic effect in vitro, even in the presence of bortezomib resistance, and modulates signaling pathways involved in MM development. CONCLUSIONS UroA might be a potential therapeutic agent to halt MM disease progression or to overcome resistance when used in combination. Video Abstract.
Collapse
Affiliation(s)
- Alba Rodríguez-García
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain.
| | - Raquel Ancos-Pintado
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain
| | - Roberto García-Vicente
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandra Ortiz-Ruiz
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Andrés Arroyo
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Miguel Ángel Navarro
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - María Luz Morales
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia Guevara-Ramirez
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Pablo Justo
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Nieves López-Muñoz
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - José Sánchez-Pina
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - Rafael Alonso
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
| | - María Victoria Selma
- Laboratory of Quality, Safety, and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain
| | | | - Rocío García-Villalba
- Laboratory of Quality, Safety, and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain
| | | | - Rosa Ayala
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
- Department of Medicine, Medicine School, Universidad Complutense de Madrid, Madrid, Spain
| | - Joaquín Martínez-López
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain
- Department of Medicine, Medicine School, Universidad Complutense de Madrid, Madrid, Spain
| | - María Linares
- Hematological Malignancies Clinical Research CRIS Unit H120-CNIO, Department of Hematology, Hospital Universitario 12 de Octubre, imas12, Universidad Complutense de Madrid, Madrid, Spain.
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
2
|
Lei K, Liang R, Liang J, Lu N, Huang J, Xu K, Tan B, Wang K, Liang Y, Wang W, Lin H, Wang M. CircPDE5A-encoded novel regulator of the PI3K/AKT pathway inhibits esophageal squamous cell carcinoma progression by promoting USP14-mediated de-ubiquitination of PIK3IP1. J Exp Clin Cancer Res 2024; 43:124. [PMID: 38658954 PMCID: PMC11040784 DOI: 10.1186/s13046-024-03054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a common gastrointestinal tumor and has become an important global health problem. The PI3K/AKT signaling pathway plays a key role in the development of ESCC. CircRNAs have been reported to be involved in the regulation of the PI3K/AKT pathway, but the underlying mechanisms are unclear. Therefore, this study aimed to identify protein-coding circRNAs and investigate their functions in ESCC. METHODS Differential expression of circRNAs between ESCC tissues and adjacent normal tissues was identified using circRNA microarray analysis. Thereafter, LC-MS/MS was used to identify circPDE5A-encoded novel protein PDE5A-500aa. Molecular biological methods were used to explore the biological functions and regulatory mechanisms of circPDE5A and PDE5A-500aa in ESCC. Lastly, circRNA-loaded nanoplatforms were constructed to investigate the therapeutic translation value of circPDE5A. RESULTS We found that circPDE5A expression was down-regulated in ESCC cells and tissues and that it was negatively associated with advanced clinicopathological stages and poorer prognosis in ESCC. Functionally, circPDE5A inhibited ESCC proliferation and metastasis in vitro and in vivo by encoding PDE5A-500aa, a key regulator of the PI3K/AKT signaling pathway in ESCC. Mechanistically, PDE5A-500aa interacted with PIK3IP1 and promoted USP14-mediated de-ubiquitination of the k48-linked polyubiquitin chain at its K198 residue, thereby attenuating the PI3K/AKT pathway in ESCC. In addition, Meo-PEG-S-S-PLGA-based reduction-responsive nanoplatforms loaded with circPDE5A and PDE5A-500aa plasmids were found to successfully inhibit the growth and metastasis of ESCC in vitro and in vivo. CONCLUSION The novel protein PDE5A-500aa encoded by circPDE5A can act as an inhibitor of the PI3K/AKT signaling pathway to inhibit the progression of ESCC by promoting USP14-mediated de-ubiquitination of PIK3IP1 and may serve as a potential target for the development of therapeutic agents.
Collapse
Affiliation(s)
- Kai Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ruihao Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jialu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Nan Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Jing Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ke Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Binghua Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kexi Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yicheng Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wenjian Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
3
|
Mangalaparthi KK, Patel K, Khan AA, Nair B, Kumar RV, Prasad TSK, Sidransky D, Chatterjee A, Pandey A, Gowda H. Molecular Characterization of Esophageal Squamous Cell Carcinoma Using Quantitative Proteomics. Cancers (Basel) 2023; 15:3302. [PMID: 37444412 DOI: 10.3390/cancers15133302] [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: 02/15/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 07/15/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a heterogeneous cancer associated with a poor prognosis in advanced stages. In India, it is the sixth most common cause of cancer-related mortality. In this study, we employed high-resolution mass spectrometry-based quantitative proteomics to characterize the differential protein expression pattern associated with ESCC. We identified several differentially expressed proteins including PDPN, TOP2A, POSTN and MMP2 that were overexpressed in ESCC. In addition, we identified downregulation of esophagus tissue-enriched proteins such as SLURP1, PADI1, CSTA, small proline-rich proteins such as SPRR3, SPRR2A, SPRR1A, KRT4, and KRT13, involved in squamous cell differentiation. We identified several overexpressed proteins mapped to the 3q24-29 chromosomal region, aligning with CNV alterations in this region reported in several published studies. Among these, we identified overexpression of SOX2, TP63, IGF2BP2 and RNF13 that are encoded by genes in the 3q26 region. Functional enrichment analysis revealed proteins involved in cell cycle pathways, DNA replication, spliceosome, and DNA repair pathways. We identified the overexpression of multiple proteins that play a major role in alleviating ER stress, including SYVN1 and SEL1L. The SYVN1/SEL1L complex is an essential part of the ER quality control machinery clearing misfolded proteins from the ER. SYVN1 is an E3 ubiquitin ligase that ubiquitinates ER-resident proteins. Interestingly, there are also other non-canonical substrates of SYVN1 which are known to play a crucial role in tumor progression. Thus, SYVN1 could be a potential therapeutic target in ESCC.
Collapse
Affiliation(s)
- Kiran K Mangalaparthi
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Krishna Patel
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
| | - Aafaque Ahmad Khan
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
| | - Rekha V Kumar
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore 560066, India
| | - Thottethodi Subrahmanya Keshav Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - David Sidransky
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Otolaryngology and Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aditi Chatterjee
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Akhilesh Pandey
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
- Manipal Academy of Higher Education, Manipal 576104, India
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560029, India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 691001, India
- Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
4
|
Xu ZJ, Liu M, Niu QJ, Huang YX, Zhao L, Lei XG, Sun LH. Both selenium deficiency and excess impair male reproductive system via inducing oxidative stress-activated PI3K/AKT-mediated apoptosis and cell proliferation signaling in testis of mice. Free Radic Biol Med 2023; 197:15-22. [PMID: 36731804 DOI: 10.1016/j.freeradbiomed.2023.01.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/02/2023]
Abstract
Selenium (Se) deficiency or excess impairs testicular development and spermatogenesis, while the underlying mechanisms in this regard remain unclear. This study was designed to explore the molecular biology of Se deficiency or excess in spermatogenesis in mice. Three-week-old male mice (n = 10 mice/diet) were fed with Se-deficient diet (SeD, 0.02 mg Se/kg), adequate-Se diet (SeA, 0.2 mg Se/kg), or excess-Se diet (SeE, 2.0 mg Se/kg) for 5 months. Compared with SeA, SeD reduced (P < 0.05) the body weight (10.4%) and sperm density (84.3%) but increased (P < 0.05) sperm deformity (32.8%); SeE decreased (P < 0.05) the sperm density (78.5%) and sperm motility (35.9%) of the mice. Meanwhile, both SeD and SeE increased (P < 0.05) serum FSH concentrations (10.4-25.6%) and induced testicular damage in mice in comparison with the SeA. Compared with SeA, SeD increased (P < 0.05) the 8-OHdG concentration by 25.5%; SeE increased (P < 0.05) both MDA and 8-OHdG concentrations by 118.8-180.3% in testis. Furthermore, transcriptome analysis showed that there 1325 and 858 transcripts were altered (P < 0.05) in the testis by SeD and SeE, respectively, compared with SeA. KEGG pathway analysis revealed that these differentially expressed genes were mainly enriched in the PI3K-AKT signaling pathway, which is regulated by oxidative stress. Moreover, western blotting analysis revealed that SeD and SeE dysregulated PI3K-AKT-mediated apoptosis and cell proliferation signaling, including upregulating (P < 0.05) caspase 3, cleaved-caspase 3, BCL-2 and (or) P53 and downregulating (P < 0.05) PI3K, p-AKT, p-mTOR, 4E-BP1, p-4E-BP1 and (or) p-p70S6K in the testis of mice compared with SeA. Additionally, compared with SeA, both SeD and SeE increased (P < 0.05) GPX3 and SELENOO; SeD decreased (P < 0.05) GPX1, TXRND3 and SELENOW, but SeE increased (P < 0.05) production of three selenoproteins in the testis. Conclusively, both Se deficiency and excess impairs male reproductive system in mice, potentially with the induction of oxidative stress and activation of PI3K/AKT-mediated apoptosis and cell proliferation signaling in the testis.
Collapse
Affiliation(s)
- Zi-Jian Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Meng Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Qin-Jian Niu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yu-Xuan Huang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
5
|
Hang Z, Wei J, Zheng M, Gui Z, Chen H, Sun L, Fei S, Han Z, Tao J, Wang Z, Tan R, Gu M. Iguratimod Attenuates Macrophage Polarization and Antibody-Mediated Rejection After Renal Transplant by Regulating KLF4. Front Pharmacol 2022; 13:865363. [PMID: 35614941 PMCID: PMC9125033 DOI: 10.3389/fphar.2022.865363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background: This study aimed to explore the effect and mechanism of iguratimod (IGT) on M1 macrophage polarization and antibody-mediated rejection (ABMR) after renal transplant.Methods: Bioinformatics analysis was performed using three public databases derived from the GEO database. Sprague–Dawley (SD) rats were pre-sensitized with donors of Wistar rats in skin transplantation and a rat renal transplant ABMR model was established from the donors to skin pre-sensitized recipients. Subsequently, IGT was treated on the ABMR model. Routine staining and immunofluorescence (IF) staining were performed to observe the pathological changes in each group and flow cytometry was performed to detect the changes of DSA titers in peripheral blood. In addition, bone-marrow-derived macrophage (BMDM) was extracted and interfered with IGT to explore the effect of IGT in vivo. PCR, IF staining, and Western blot were used to detect the expression of related genes and proteins.Results: Bioinformatics analysis revealed that several immune cells were significantly infiltrated in the ABMR allograft, while M1 macrophage was noticed with the most significance. Results of IF staining and PCR proved the findings of the bioinformatics analysis. Based on this, IGT was observed to significantly attenuate the degree of peritubular capillary vasculitis and arteriolitis in the rat renal transplant ABMR model, whereas it decreases the expression of C4d and reduces the titer of DSA. Results in vitro suggested that M1 macrophage-related transcripts and proteins were significantly reduced by the treatment of IGT in a dose- and time-dependent manner. Furthermore, IGT intervention could remarkably decrease the expression of KLF4.Conclusion: Polarization of M1 macrophages may aggravate ABMR after renal transplant by promoting DSA-mediated endothelial cell injury, and IGT may attenuate the pathogenesis of ABMR by targeting KLF4.
Collapse
Affiliation(s)
- Zhou Hang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jintao Wei
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Zheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zeping Gui
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Sun
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Fei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zijie Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| | - Min Gu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zijie Wang, ; Min Gu, ; Ruoyun Tan,
| |
Collapse
|
6
|
Huang R, Dai Q, Yang R, Duan Y, Zhao Q, Haybaeck J, Yang Z. A Review: PI3K/AKT/mTOR Signaling Pathway and Its Regulated Eukaryotic Translation Initiation Factors May Be a Potential Therapeutic Target in Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:817916. [PMID: 35574327 PMCID: PMC9096244 DOI: 10.3389/fonc.2022.817916] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/01/2022] [Indexed: 11/15/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignant tumor developing from the esophageal squamous epithelium, and is the most common histological subtype of esophageal cancer (EC). EC ranks 10th in morbidity and sixth in mortality worldwide. The morbidity and mortality rates in China are both higher than the world average. Current treatments of ESCC are surgical treatment, radiotherapy, and chemotherapy. Neoadjuvant chemoradiotherapy plus surgical resection is recommended for advanced patients. However, it does not work in the significant promotion of overall survival (OS) after such therapy. Research on targeted therapy in ESCC mainly focus on EGFR and PD-1, but neither of the targeted drugs can significantly improve the 3-year and 5-year survival rates of disease. Phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway is an important survival pathway in tumor cells, associated with its aggressive growth and malignant progression. Specifically, proliferation, apoptosis, autophagy, and so on. Related genetic alterations of this pathway have been investigated in ESCC, such as PI3K, AKT and mTOR-rpS6K. Therefore, the PI3K/AKT/mTOR pathway seems to have the capability to serve as research hotspot in the future. Currently, various inhibitors are being tested in cells, animals, and clinical trials, which targeting at different parts of this pathway. In this work, we reviewed the research progress on the PI3K/AKT/mTOR pathway how to influence biological behaviors in ESCC, and discussed the interaction between signals downstream of this pathway, especially eukaryotic translation initiation factors (eIFs) and the development and progression of ESCC, to provide reference for the identification of new therapeutic targets in ESCC.
Collapse
Affiliation(s)
- Ran Huang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qiong Dai
- Department of Human Anatomy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ruixue Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Duan
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qi Zhao
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Luo Q, Du R, Liu W, Huang G, Dong Z, Li X. PI3K/Akt/mTOR Signaling Pathway: Role in Esophageal Squamous Cell Carcinoma, Regulatory Mechanisms and Opportunities for Targeted Therapy. Front Oncol 2022; 12:852383. [PMID: 35392233 PMCID: PMC8980269 DOI: 10.3389/fonc.2022.852383] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC), is the most common type of esophageal cancer worldwide, mainly occurring in the Asian esophageal cancer belt, including northern China, Iran, and parts of Africa. Phosphatidlinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway is one of the most important cellular signaling pathways, which plays a crucial role in the regulation of cell growth, differentiation, migration, metabolism and proliferation. In addition, mutations in some molecules of PI3K/Akt/mTOR pathway are closely associated with survival and prognosis in ESCC patients. A large number of studies have found that there are many molecules in ESCC that can regulate the PI3K/Akt/mTOR pathway. Overexpression of these molecules often causes aberrant activation of PI3K/Akt/mTOR pathway. Currently, several effective PI3K/Akt/mTOR pathway inhibitors have been developed, which can play anticancer roles either alone or in combination with other inhibitors. This review mainly introduces the general situation of ESCC, the composition and function of PI3K/Akt/mTOR pathway, and regulatory factors that interact with PI3K/Akt/mTOR signaling pathway. Meanwhile, mutations and inhibitors of PI3K/Akt/mTOR pathway in ESCC are also elucidated.
Collapse
Affiliation(s)
- Qian Luo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Ruijuan Du
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Wenting Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Guojing Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.,Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Zhang K, Chai B, Ji H, Chen L, Ma Y, Zhu L, Xu J, Wu Y, Lan Y, Li H, Feng Z, Xiao J, Zhang H, Xu K. Bioglass promotes wound healing by inhibiting endothelial cell pyroptosis through regulation of the connexin 43/reactive oxygen species (ROS) signaling pathway. J Transl Med 2022; 102:90-101. [PMID: 34521991 DOI: 10.1038/s41374-021-00675-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/22/2021] [Accepted: 08/30/2021] [Indexed: 11/08/2022] Open
Abstract
Bioactive glass (BG) has recently shown great promise in soft tissue repair, especially in wound healing; however, the underlying mechanism remains unclear. Pyroptosis is a novel type of programmed cell death that is involved in various traumatic injury diseases. Here, we hypothesized that BG may promote wound healing through suppression of pyroptosis. To test this scenario, we investigated the possible effect of BG on pyroptosis in wound healing both in vivo and in vitro. This study showed that BG can accelerate wound closure, granulation formation, collagen deposition, and angiogenesis. Moreover, western blot analysis and immunofluorescence staining revealed that BG inhibited the expression of pyroptosis-related proteins in vivo and in vitro. In addition, while BG regulated the expression of connexin43 (Cx43), it inhibited reactive oxygen species (ROS) production. Cx43 activation and inhibition experiments further indicate that BG inhibited pyroptosis in endothelial cells by decreasing Cx43 expression and ROS levels. Taken together, these studies suggest that BG promotes wound healing by inhibiting pyroptosis via Cx43/ROS signaling pathway.
Collapse
Affiliation(s)
- Kailun Zhang
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Bo Chai
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Hao Ji
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Liuqing Chen
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Yanbing Ma
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Lifei Zhu
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Jingyu Xu
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Yanqing Wu
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China
| | - Yinan Lan
- Department of Orthopedic Surgery, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Hao Li
- Department of Orthopedics Surgery, Lishui People's Hospital, The sixth affiliated hospital of Wenzhou medical university, Lishui, Zhejiang, China
| | - Zhiguo Feng
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Ke Xu
- Institute of Life Sciences, Engineering Laboratory of Zhejiang province for pharmaceutical development of growth factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Zhejiang, China.
| |
Collapse
|
9
|
Zuo Y, Yi L, Lu S. Dermal fibroblast from superficial layers of pig skin exhibits more proliferative capacity than that from deep layers. J Tissue Viability 2021; 31:278-285. [PMID: 34895969 DOI: 10.1016/j.jtv.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To further examine the feasibility of using pigs as an animal model for the study of dermal fibroblast heterogeneity and to explore the proliferative capacity of dermal fibroblasts from different layers of pig skin in vitro and in vivo. MATERIAL AND METHODS Cultured superficial and deep dermal fibroblasts were subjected to cell growth assay, cell cycle analysis, immunocytochemical staining and western blotting for proliferating cell nuclear antigens. Moreover, skin samples autografted with superficial/deep dermal fibroblasts were subjected to immunohistochemical staining and western blotting for proliferating cell nuclear antigen. RESULTS The cell growth assay showed that the growth curve of the superficial dermal fibroblast was progressively higher than that of the deep layer. The cell cycle analysis showed that the (G2+S) percentage of the superficial dermal fibroblasts was significantly higher than that of the deep layer fibroblasts. The immunocytochemical staining and western blotting showed that the expression of proliferating cell nuclear antigen in the cultured superficial dermal fibroblast was significantly higher than that of the deep layer cells. The immunohistochemical staining showed that the positive rate of proliferating cell nuclear antigen in the skin samples autografted with the superficial dermal fibroblast was significantly higher than that of the deep layer. CONCLUSIONS This study has demonstrated that similar to human dermal fibroblasts, dermal fibroblasts from different layers of pig skin exhibit distinct proliferative capacity, which increases the feasibility of using pigs as an animal model for future studies on the heterogeneity of dermal fibroblasts.
Collapse
Affiliation(s)
- Yanhai Zuo
- Department of Orthopedics, SiJing Hospital of SongJiang District, Shanghai, China; Wound Healing Center, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lei Yi
- Department of Burn, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Shuliang Lu
- Wound Healing Center, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
10
|
Niu Z, Li Y, Xu Y, Jiang W, Tao R, Chen Y, Han Y. Silencing FYVE, RhoGEF, and PH domain containing 1 (FGD1) suppresses melanoma progression by inhibiting PI3K/AKT signaling pathway. Bioengineered 2021; 12:12193-12205. [PMID: 34783295 PMCID: PMC8810171 DOI: 10.1080/21655979.2021.2005877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
Cutaneous melanoma is the leading cause of death among skin cancers despite the availability of diverse treatments. FGD1 plays an important role in multiple cancers, but how it works in cutaneous melanoma has not been illustrated. Thus, this study was intended to investigate the roles of FGD1 and its underlying mechanisms in cutaneous melanoma. Bioinformatics tools and quantitative real-time polymerase chain reaction (qRT-PCR) were used to analyze the expression of FGD1 in cutaneous melanoma. After the knockdown of FGD1 in melanoma cells, the proliferation, migration, and invasion of cells were analyzed by cell counting kit-8 (CCK8) assay, colony formation assays and transwell assays. Western blot was used to check the expression of key factors in PI3K/AKT pathway. In addition, nude mice models were used to study the role of FGD1 in melanoma development and metastasis in vivo. The data demonstrated that FGD1 was up-regulated and predicted a poor clinical outcome for cutaneous melanoma patients. Knockdown of FGD1 inhibited melanoma cell proliferation, migration, and invasion. The expressions of p-PI3K and p-AKT were significantly decreased, while the expressions of PI3K and AKT showed no marked difference in the knockdown group. Meanwhile, knockdown of FGD1 suppressed the development of melanoma in vivo. This study suggested that knockdown of FGD1 could block melanoma formation and proliferation by inhibiting PI3K/AKT signaling pathway. FGD1 might be a promising therapeutic target for melanoma.
Collapse
Affiliation(s)
- Zehao Niu
- Medical School of Chinese PLA, Beijing, China
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Li
- Medical School of Chinese PLA, Beijing, China
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yujian Xu
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Weiqian Jiang
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Youbai Chen
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Lu X, Liu M, Liao Y, Huang C, Chai L, Jin Y, Xiong Q, Chen B. Meta-analysis of the association between mTORC1-related genes polymorphisms and cancer risk. Pathol Res Pract 2021; 229:153696. [PMID: 34839094 DOI: 10.1016/j.prp.2021.153696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND mTOR, mLST8 and RAPTOR are the core components of mTORC1, which has been found to be closely related to tumorigenesis. Currently, multiple single nucleotide polymorphisms (SNPs) in mTOR gene (rs2295080, rs17036508 and rs1034528), mLST8 gene (rs3160 and rs26865) and RPTOR gene (rs1062935, rs3751932, rs3751834, rs12602885) have been extensively studied for their associations with cancer risk. However, the results remained inconclusive and conflicting. Therefore, we here performed a meta-analysis of all available studies to investigate the association between these SNPs and cancer risk. METHODS Up to April 2021, 25 related publications were retrieved and included in this meta-analysis. The odds ratios (ORs) and 95% confidence intervals (CIs) calculated by fixed or random effects models were applied to assess the strength of association. Trial Sequential Analysis (TSA) was conducted to weaken the random error and enhance the reliability of evidence. RESULTS After Bonferroni correction, it was revealed that rs3160, rs26865, rs1062935, rs3751932, rs3751834 and rs10602885 were not associated with cancer risk. However, rs17036508 and rs1034528 showed significant association with total cancer risk. A significant association was also found between rs2295080 and total cancer risk, and stratified analysis by cancer type suggested that rs2295080 was specifically associated with acute lymphoblastic leukemia risk, prostate cancer risk, and breast cancer risk. CONCLUSIONS The present meta-analysis suggested that the rs2295080, rs17036508 and rs1034528 polymorphisms in mTOR gene may be the susceptive factors for cancer development, while the target genetic polymorphisms in mLST8 gene or RPTOR gene may not be associated with cancer risk. However, these findings remain to be confirmed or further reinforced in large and well-designed studies in different ethnic populations.
Collapse
Affiliation(s)
- Xiaoling Lu
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Meitong Liu
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Yuxiao Liao
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Chao Huang
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Longlong Chai
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China; Department of Laboratory, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Yuchen Jin
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Qiantao Xiong
- Department of Laboratory, Maternal and Child Health Hospital of Hubei Province, Wuhan, China.
| | - Bifeng Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China.
| |
Collapse
|
12
|
Zhu DS, Dong JY, Xu YY, Zhang XT, Fu SB, Liu W. Omipalisib Inhibits Esophageal Squamous Cell Carcinoma Growth Through Inactivation of Phosphoinositide 3-Kinase (PI3K)/AKT/Mammalian Target of Rapamycin (mTOR) and ERK Signaling. Med Sci Monit 2020; 26:e927106. [PMID: 32804918 PMCID: PMC7450785 DOI: 10.12659/msm.927106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a life-threatening digestive tract malignancy with no known curative treatment. This study aimed to investigate the antineoplastic effects of omipalisib and its underlying molecular mechanisms in ESCC using a high throughput screen. Material/Methods MTT assay and clone formation were used to determine cell viability and proliferation. Flow cytometry was conducted to detect cell cycle distribution and apoptosis. Global gene expression and mRNA expression levels were determined by RNA sequencing and real-time PCR, respectively. Protein expression was evaluated in the 4 ESCC cell lines by Western blot analysis. Finally, a xenograft nude mouse model was used to evaluate the effect of omipalisib on tumor growth in vivo. Results In the pilot screening of a 1404-compound library, we demonstrated that omipalisib markedly inhibited cell proliferation in a panel of ESCC cell lines. Mechanistically, omipalisib induced G0/G1 cell cycle arrest and apoptosis. RNA-seq, KEGG, and GSEA analyses revealed that the PI3K/AKT/mTOR pathway is the prominent target of omipalisib in ESCC cells. Treatment with omipalisib decreased expression of p-AKT, p-4EBP1, p-p70S6K, p-S6, and p-ERK, therefore disrupting the activation of PI3K/AKT/mTOR and ERK signaling. In the nude mouse xenograft model, omipalisib significantly suppressed the tumor growth in ESCC tumor-bearing mice without obvious adverse effects. Conclusions Omipalisib inhibited the proliferation and growth of ESCC by disrupting PI3K/AKT/mTOR and ERK signaling. The present study supports the rationale for using omipalisib as a therapeutic approach in ESCC patients. Further clinical studies are needed.
Collapse
Affiliation(s)
- Dong-Shan Zhu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Jing-Yao Dong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Yao-Yao Xu
- The Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Xin-Tong Zhang
- The Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Shi-Bo Fu
- The Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
13
|
Lu Z, Shi X, Gong F, Li S, Wang Y, Ren Y, Zhang M, Yu B, Li Y, Zhao W, Zhang J, Hou G. RICTOR/mTORC2 affects tumorigenesis and therapeutic efficacy of mTOR inhibitors in esophageal squamous cell carcinoma. Acta Pharm Sin B 2020; 10:1004-1019. [PMID: 32642408 PMCID: PMC7332809 DOI: 10.1016/j.apsb.2020.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/01/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of mTORC1/mTORC2 pathway is observed in many cancers and mTORC1 inhibitors have been used clinically in many tumor types; however, the mechanism of mTORC2 in tumorigenesis is still obscure. Here, we mainly explored the potential role of mTORC2 in esophageal squamous cell carcinoma (ESCC) and its effects on the sensitivity of cells to mTOR inhibitors. We demonstrated that RICTOR, the key factor of mTORC2, and p-AKT (Ser473) were excessively activated in ESCC and their overexpression is related to lymph node metastasis and the tumor-node-metastasis (TNM) phase of ESCC patients. Furthermore, we found that mTORC1/ mTORC2 inhibitor PP242 exhibited more efficacious anti-proliferative effect on ESCC cells than mTORC1 inhibitor RAD001 due to RAD001-triggered feedback activation of AKT signal. Another, we demonstrated that down-regulating expression of RICTOR in ECa109 and EC9706 cells inhibited proliferation and migration as well as induced cell cycle arrest and apoptosis. Noteworthy, knocking-down stably RICTOR significantly suppresses RAD001-induced feedback activation of AKT/PRAS40 signaling, and enhances inhibition efficacy of PP242 on the phosphorylation of AKT and PRAS40, thus potentiates the antitumor effect of RAD001 and PP242 both in vitro and in vivo. Our findings highlight that selective targeting mTORC2 could be a promising therapeutic strategy for future treatment of ESCC.
Collapse
Key Words
- 4EBP-1, E binding protein-1
- AKT
- AKT, protein kinase B (PKB)
- ESCC, esophageal squamous cell carcinoma
- Esophageal squamous cell carcinoma
- FDA, U.S. Food and Drug Administration
- H&E staining, hematoxylin and eosin staining
- IC50, half maximal inhibitory concentration
- PI3K, phosphatidylinositol 3 kinase
- RAD001
- RICTOR
- RICTOR, rapamycin-insensitive companion of mTOR
- TNM, tumor-node-metastasis
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- mTOR, mammalian target of rapamycin
- mTORC1, mTOR complex 1
- mTORC2, mTOR complex 2
- p70S6K, p70 ribosomal S6 kinase-1
- pp242
- rapalogs, rapamycin and its analogs
Collapse
|
14
|
Zhong W, Tang X, Liu Y, Zhou C, Liu P, Li E, Zhong P, Lv H, Zou Q, Wang M. Benzoxazole Derivative K313 Induces Cell Cycle Arrest, Apoptosis and Autophagy Blockage and Suppresses mTOR/p70S6K Pathway in Nalm-6 and Daudi Cells. Molecules 2020; 25:molecules25040971. [PMID: 32098126 PMCID: PMC7070478 DOI: 10.3390/molecules25040971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Benzoxazole derivative K313 has previously been reported to possess anti-inflammatory effects in lipopolysaccharide-induced RAW264.7 macrophages. To date, there have been no related reports on the anticancer effects of K313. In this study, we found that K313 reduced the viability of human B-cell leukemia (Nalm-6) and lymphoma (Daudi) cells in a dose-dependent manner without affecting healthy peripheral blood mononuclear cells (PBMCs) and induced moderate cell cycle arrest at the G0/G1 phase. Meanwhile, K313 mediated cell apoptosis, which was accompanied by the activation of caspase-9, caspase-3, and poly ADP-ribose polymerase (PARP). Furthermore, cells treated with K313 showed a significant decrease in mitochondrial membrane potential (MMP), which may have been caused by the caspase-8-mediated cleavage of Bid, as detected by Western blot analysis. We also found that K313 led to the downregulation of p-p70S6K protein, which plays an important role in cell survival and cell cycle progression. In addition, treatment of these cells with K313 blocked autophagic flux, as reflected in the accumulation of LC3-II and p62 protein levels in a dose- and time-dependent manner. In conclusion, K313 decreases cell viability without affecting normal healthy PBMCs, induces cell cycle arrest and apoptosis, reduces p-p70S6K protein levels, and mediates strong autophagy inhibition. Therefore, K313 and its derivatives could be developed as potential anticancer drugs or autophagy blockers in the future.
Collapse
Affiliation(s)
- Wenying Zhong
- Key Laboratory of Bio-resources and Eco-environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China;
| | - Xinwen Tang
- Center of Science and Research, Chengdu Medical College, Chengdu 610513, China; (X.T.); (Y.L.)
| | - Yang Liu
- Center of Science and Research, Chengdu Medical College, Chengdu 610513, China; (X.T.); (Y.L.)
| | - Chunyu Zhou
- School of Pharmacy, Chengdu Medical College, Chengdu 610083, China; (C.Z.); (E.L.); (P.Z.); (H.L.)
| | - Pan Liu
- College of Biological Science and Technology, Chengdu Medical College, Chengdu 610500, China;
| | - Enhui Li
- School of Pharmacy, Chengdu Medical College, Chengdu 610083, China; (C.Z.); (E.L.); (P.Z.); (H.L.)
| | - Peilin Zhong
- School of Pharmacy, Chengdu Medical College, Chengdu 610083, China; (C.Z.); (E.L.); (P.Z.); (H.L.)
| | - Haoxue Lv
- School of Pharmacy, Chengdu Medical College, Chengdu 610083, China; (C.Z.); (E.L.); (P.Z.); (H.L.)
| | - Qiang Zou
- Center of Science and Research, Chengdu Medical College, Chengdu 610513, China; (X.T.); (Y.L.)
- Correspondence: (Q.Z.); (M.W.); Tel.: +86-028-62739159 (Q.Z.); +86-028-85418776 (M.W.)
| | - Maolin Wang
- Key Laboratory of Bio-resources and Eco-environment of the Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China;
- Correspondence: (Q.Z.); (M.W.); Tel.: +86-028-62739159 (Q.Z.); +86-028-85418776 (M.W.)
| |
Collapse
|
15
|
SOX9/miR-203a axis drives PI3K/AKT signaling to promote esophageal cancer progression. Cancer Lett 2020; 468:14-26. [DOI: 10.1016/j.canlet.2019.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 09/22/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
|
16
|
Liposome-based co-delivery of 7-O-geranyl-quercetin and IGF-1R siRNA for the synergistic treatment of non-small cell lung cancer. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Javadinia SA, Shahidsales S, Fanipakdel A, Mostafapour A, Joudi-Mashhad M, Ferns GA, Avan A. The Esophageal Cancer and the PI3K/AKT/mTOR Signaling Regulatory microRNAs: a Novel Marker for Prognosis, and a Possible Target for Immunotherapy. Curr Pharm Des 2019; 24:4646-4651. [PMID: 30636576 DOI: 10.2174/1381612825666190110143258] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 01/25/2023]
Abstract
The Phosphatidylinositol 3-kinase/AKT/Mammalian Target of Rapamycin (PI3K/AKT/mTOR) pathway has a critical regulatory role in cell biology including translation, transcription, and autophagy. Dysregulation of this pathway is involved in the pathogenesis, development, and prognosis of esophageal cancer that has been assessed in the recent years and its potential as a target in therapy. This report summarizes the current knowledge about PI3K/AKT/mTOR pathway and its cross-talk with a focus on the value of targeting this pathway as a potential therapeutic target in the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Seyed A Javadinia
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Azar Fanipakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mostafapour
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Joudi-Mashhad
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Department of Medical Education, Brighton and Sussex Medical School Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, United Kingdom
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Chamcheu JC, Roy T, Uddin MB, Banang-Mbeumi S, Chamcheu RCN, Walker AL, Liu YY, Huang S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019; 8:cells8080803. [PMID: 31370278 PMCID: PMC6721560 DOI: 10.3390/cells8080803] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) and associated phosphatidyl-inositiol 3-kinase (PI3K)/protein kinase B (Akt) pathways regulate cell growth, differentiation, migration, and survival, as well as angiogenesis and metabolism. Dysregulation of these pathways is frequently associated with genetic/epigenetic alterations and predicts poor treatment outcomes in a variety of human cancers including cutaneous malignancies like melanoma and non-melanoma skin cancers. Recently, the enhanced understanding of the molecular and genetic basis of skin dysfunction in patients with skin cancers has provided a strong basis for the development of novel therapeutic strategies for these obdurate groups of skin cancers. This review summarizes recent advances in the roles of PI3K/Akt/mTOR and their targets in the development and progression of a broad spectrum of cutaneous cancers and discusses the current progress in preclinical and clinical studies for the development of PI3K/Akt/mTOR targeted therapies with nutraceuticals and synthetic small molecule inhibitors.
Collapse
Affiliation(s)
| | - Tithi Roy
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Sergette Banang-Mbeumi
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | | | - Anthony L Walker
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Yong-Yu Liu
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
19
|
Mo Y, Wang Y, Xiong F, Ge X, Li Z, Li X, Li Y, Li X, Xiong W, Li G, Zeng Z, Guo C. Proteomic Analysis of the Molecular Mechanism of Lovastatin Inhibiting the Growth of Nasopharyngeal Carcinoma Cells. J Cancer 2019; 10:2342-2349. [PMID: 31258737 PMCID: PMC6584415 DOI: 10.7150/jca.30454] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Metabolic abnormalities are one of the essential features of tumors. Increasingly more studies have shown that lovastatin, a lipid-reducing drug, has visible inhibitory effects on tumors, but it has not been reported in nasopharyngeal carcinoma. In this paper, we explored the effects of lovastatin on the growth of nasopharyngeal carcinoma cells and its possible molecular mechanisms. After treating nasopharyngeal carcinoma cells with different concentrations of lovastatin, we found that lovastatin can inhibit the growth of nasopharyngeal carcinoma in a time- and dose-dependent manner. To explore the molecular mechanism of how lovastatin inhibits the growth of nasopharyngeal carcinoma, we examined the proteome of nasopharyngeal carcinoma cells treated at different time points using an LC/MS whole-proteomic strategy. The molecular network of differentially expressed proteins was constructed using IPA software. It was found that lovastatin inhibited the growth of nasopharyngeal carcinoma cells mainly by affecting the EIF2 and the mTOR pathways, which regulate cell metabolism and apoptosis. The results of this study provide a robust basis for further research on the molecular mechanism of lovastatin's inhibition of nasopharyngeal carcinoma cells and provide a reference for the clinical use of lovastatin in the treatment of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Xiaolu Ge
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Zheng Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis, Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410078, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
20
|
Zhang Q, Yan L, Chen M, Gui M, Lu L, Deng F, Ren Z. IgA1 isolated from Henoch-Schönlein purpura children promotes proliferation of human mesangial cells in vitro. Cell Biol Int 2019; 43:760-769. [PMID: 30958627 DOI: 10.1002/cbin.11142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/23/2019] [Indexed: 01/31/2023]
Abstract
Previous studies show that the proliferation of human mesangial cells (HMCs) played a significant part in the pathogenesis of Henoch-Schönlein purpura nephritis (HSPN). The aim of this study was to explore the proliferation of HMCs induced by IgA1 isolated from the sera of HSP patients. HMCs were cultured in three different types of media, including IgA1 from patients with HSP (HSP IgA1 group), healthy children (healthy IgA1 group) and medium (control group). The proliferation of HMCs incubated with IgA1 was determined by cell counting kit-8 assay and bromodeoxyuridine incorporation. The expression of ERK1/2 and phosphatidylinositol 3 kinase/protein kinase B/mammalian targets of the rapamycin (PI3K/AKt/mTOR) signals and transferrin receptor (TfR/CD71) was detected with the methods of immunoblotting. The results indicated that the proliferation of HMCs significantly increased in the HSP IgA1 group compared with that in the control group or the healthy IgA1 group (P < 0.001). Moreover, we found that IgA1 isolated from HSP patients activated ERK and PI3K/AKt/mTOR signals, and markedly increased TfR/CD71 expression in HMCs. These effects induced by IgA1 isolated from patients with HSP were inhibited by human TfR polyclonal antibody (hTfR pAb) and soluble human transferrin receptor (sTfR), indicating that IgA1-induced HMC proliferation and ERK1/2 and PI3K/AKt/mTOR activation were dependent on TfR/CD71 engagement. Altogether, these data suggested that TfR/CD71 overexpression and ERK1/2 and PI3K/AKt/mTOR activation were engaged in HMC proliferation induced by IgA1 from HSP patients, which might be related to the mesangial injury of HSPN.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Heifei, Anhui, China
| | - Lili Yan
- Department of Anatomy, Anhui Medical University, Hefei, Anhui, China
| | - Mingyu Chen
- Department of Anatomy, Anhui Medical University, Hefei, Anhui, China
| | - Ming Gui
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Heifei, Anhui, China
| | - Ling Lu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Heifei, Anhui, China
| | - Fang Deng
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Heifei, Anhui, China
| | - Zhenhua Ren
- Department of Anatomy, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
21
|
Zuo J, Jiang Y, Zhang E, Chen Y, Liang Z, Zhu J, Zhao Y, Xu H, Liu G, Liu J, Wang W, Zhang S, Zhen Y. Synergistic effects of 7-O-geranylquercetin and siRNAs on the treatment of human breast cancer. Life Sci 2019; 227:145-152. [PMID: 31009625 DOI: 10.1016/j.lfs.2019.04.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 10/27/2022]
Abstract
AIMS To investigate the antitumor effect of 7-O-geranylquercetin (GQ) combining with survivin siRNA (siSuvi) or IL-10 siRNA (siIL-10) to breast cancer. MAIN METHODS Xenograft tumor model was established by subcutaneously inoculating human breast cancer MCF-7 cells in BALB/c nude mice. Transfection efficiency of siRNA mediated by cationic liposome CDO14 in MCF-7 cells and tumor bearing mice was measured by flow cytometer and living imaging sysytem, respectively. Cell viability was detected using CCK-8 assay. Cell apoptosis was determined by Hoechst33342 staining and AV-PI staining. Tumors bearing mice were administered with GQ by gavage, and/or with liposome CDO14 mediated siRNAs via tail intravenous injection. Expression levels of proteins and cytokines were detected by western blot and ELISA, respectively. KEY FINDINGS Liposome CDO14 could deliver siRNA to tumor effectively. Combination of GQ and siSuvi promoted the antiproliferation and pro-apoptosis effects of GQ or siSuvi to MCF-7 cells, and reduced the level of survivin and raised the level of caspase-7 in cells. GQ combining with siSuvi inhibited the growth of tumor, down-regulated the expression of survivin and up-regulated the expression of caspase-7 in tumor tissue. Similarly, GQ combining with siIL-10 inhibited the growth of tumor, decreased the level of IL-10 and increased the level of TNF-α. These results revealed that GQ enhanced the pro-apoptosis effect of siSuvi on tumor cells and the modulating effect of siIL-10 on tumor microenvironment. SIGNIFICANCES Synergistic anti-tumor effect of GQ and siRNAs against breast cancer proved that chemical drugs combining with siRNAs is a promising antitumor strategy.
Collapse
Affiliation(s)
- Jiaxin Zuo
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yameng Jiang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Enxia Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yuling Chen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jie Zhu
- Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Hong Xu
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guoliang Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jiasi Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Wei Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
22
|
Toxicological exploration of peptide-based cationic liposomes in siRNA delivery. Colloids Surf B Biointerfaces 2019; 179:66-76. [PMID: 30947085 DOI: 10.1016/j.colsurfb.2019.03.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/05/2019] [Accepted: 03/24/2019] [Indexed: 01/22/2023]
Abstract
The toxicology of cationic liposomes was explored to advance clinical trials of liposome-mediated gene therapy through the analysis of a peptide cationic liposome with DOTAP as a positive control. We first investigated the delivery of luciferase siRNA by several peptide liposomes in mice bearing lung cancer A549 cell xenografts. Of these, a cationic liposome (CDO14) was selected for further investigation. CDO14 efficiently mediated IGF-1R-siRNA delivery and inhibited the growth of the A549 cell xenografts. The in vivo toxicity and toxicological mechanisms of the selected liposome were evaluated to assess its potential utility for gene delivery. Specifically, the effects of CDO14 on mouse body weight, hematology, urine, serum biochemical indices, and histopathology were measured in acute toxicity and subchronic toxicity tests. CDO14 showed limited toxicological effects at low dosages although it induced pulmonary inflammation and liver injury at higher dosages. The toxicity of CDO14 was lower than that of DOTAP, and the toxicity of CDO14 did not change when complexed with siRNA. The pulmonary inflammation induced by CDO14 occurred via expressional up-regulation of the pro-inflammatory cytokines TNF-α and IL-6, and expressional down-regulation of the anti-inflammatory cytokine IL-10. Liver injury induced by CDO14 was mediated by the JAK2-STAT3 signaling pathway. Lastly, CDO14 did not affect the expression of apoptosis-related proteins in normal liver cells, suggesting that it did not induce apoptosis of normal cells. The toxicological results demonstrate that peptide-based headgroups in lipids are superior to those with quaternary ammonium headgroups that are used as gene vectors for cancer therapy.
Collapse
|
23
|
Gu Y, Lu J, Sun W, Jin R, Ohira T, Zhang Z, Tian X. Oxymatrine and its metabolite matrine contribute to the hepatotoxicity induced by radix Sophorae tonkinensis in mice. Exp Ther Med 2019; 17:2519-2528. [PMID: 30906440 PMCID: PMC6425122 DOI: 10.3892/etm.2019.7237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
Previous studies by our group demonstrated that radix Sophorae tonkinensis could induce hepatotoxicity. However, it remains unclear which components of this herb may be responsible for its hepatotoxicity. The present study aimed to investigate the hepatic toxicity of treatment with matrine (MT) and oxymatrine (OMT) alone or simultaneously. Furthermore, the current study aimed to identify whether the hepatotoxicity induced by OMT is actually the toxic characterization of its metabolite MT. Hepatotoxicity was evaluated by biochemical and histopathological approaches in subchronic toxicity in mice, as well as via evaluation of cytotoxicity and enzyme leakage in AML12 liver cells. The results indicated that treatment of mice with OMT and MT individually or simultaneously resulted in centrilobular hypertrophy in the liver at doses equivalent to that contained in radix S. tonkinensis at a hepatotoxic dose, suggesting that MT and OMT are likely hepatotoxic components of this herb. OMT-induced hepatotoxicity may be primarily exerted via its metabolite MT in mice. Furthermore, OMT combined with MT was observed to be more toxic compared with OMT or MT alone. These results extend our understanding of the hepatotoxicity of radix S. tonkinensis and its active ingredients.
Collapse
Affiliation(s)
- Yingmin Gu
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jinyao Lu
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Wei Sun
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Ruomin Jin
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Toko Ohira
- Shanghai Innostar Biotech Co., Ltd., China National Shanghai Center for New Drug Safety Evaluation and Research, Shanghai 201203, P.R. China
| | - Zean Zhang
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xuesong Tian
- Center for Drug Safety Evaluation and Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China.,Center for Laboratory Animals, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
24
|
Xu S, Hui L, Yang N, Wang Y, Zhao N, Jiang XJ. Upregulation of microRNA‑194‑5p inhibits hypopharyngeal carcinoma cell proliferation, migration and invasion by targeting SMURF1 via the mTOR signaling pathway. Int J Oncol 2019; 54:1245-1255. [PMID: 30720112 PMCID: PMC6411369 DOI: 10.3892/ijo.2019.4711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 11/23/2018] [Indexed: 12/25/2022] Open
Abstract
Hypopharyngeal carcinoma (HPC) is an aggressive malignancy with the worst prognosis among all head and neck cancers. MicroRNAs (miRNAs) are involved in the development of many human cancers, and may function as oncogenes or tumor suppressors. The present study aimed to evaluate the effects of miRNA (miR)-194-5p on the proliferation and invasion of HPC cells and to identify the potential regulatory mechanism. First, miR-194-5p and Smad ubiquitin regulatory factor 1 (SMURF1) expression levels were examined in HPC tissues. Subsequently, to explore the effects of miR-194-5p on SMURF1, a dual-luciferase reporter gene assay was performed to verify the target relationship. To define the role of miR-194-5p in HPC progression, miR-194-5p upregulation and depletion were used to evaluate its effects on cell viability, invasion and migration. SMURF1 silencing and rapamycin [an inhibitor of the mammalian target of rapamycin (mTOR) signaling pathway] treatment were also used to analyze the regulatory mechanism in HPC. Finally, tumor growth was assessed in xenografted tumors in nude mice. SMURF1 was demonstrated to be highly expressed, whereas miR-194-5p was poorly expressed in HPC tissues; SMURF1 was identified as a target gene of miR-194-5p. FaDu hypopharyngeal squamous cell carcinoma cells treated with miR-194-5p mimics exhibited decreased viability, invasion and migration. The results indicated that miR-194-5p may inactivate the mTOR signaling pathway by targeting SMURF1. In addition, the in vivo experiments further verified these regulatory effects. These data suggested that miR-194-5p-targeted SMURF1 inhibition may be involved in the disruption of HPC progression through the repression of the mTOR signaling pathway.
Collapse
Affiliation(s)
- Shan Xu
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lian Hui
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Yang
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yan Wang
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ning Zhao
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xue-Jun Jiang
- Department of Ear, Nose and Throat, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
25
|
Cui Y, Liu X, Li S, Hao L, Du J, Gao D, Kang Q, Lu J. Extraction, characterization and biological activity of sulfated polysaccharides from seaweed Dictyopteris divaricata. Int J Biol Macromol 2018; 117:256-263. [PMID: 29792963 DOI: 10.1016/j.ijbiomac.2018.05.134] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/15/2018] [Accepted: 05/19/2018] [Indexed: 12/15/2022]
Abstract
Dictyopteris divaricata is a kind of important brown algae with many biological activities. It has been receiving more and more attention, yet there are rarely studies done on its polysaccharides. In this study, the optimum extraction and biological activity of seaweed polysaccharides from Dictyopteris divaricata (DDSP) were investigated. Response surface methodology (RSM), based on a three-level, three-variable Box-Behnken design (BBD), was employed to obtain the best possible combinations for maximum polysaccharides yield. The optimum extraction conditions were as follows: liquid-solid ratio of 110 mL/g, extraction time of 6 h and extraction temperature of 100 °C. Under these conditions, the experimental yield was 3.05%, which was in close agreement with the predicted value of 3.15%. The average molecular weight of DDSP was 58.05 kDa. Gas chromatograph (GC) results showed that DDSP was composed of fucose, xylose, mannose, glucose and galactose with the corresponding molar ratio of 4.45:2.74:1.00:2.94:1.35. Biological activity showed that DDSP exhibited strong antioxidant activity in vitro and possessed the potential on stimulating immune response of RAW264.7 cells. So DDSP can be used as a natural ingredient in functional foods.
Collapse
Affiliation(s)
- Yinxin Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Shufang Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Limin Hao
- The Quartermaster Research Institute of Engineering and Technology, Academy of Military Sciences PLA China, Beijing 100010, China
| | - Juan Du
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - DaHai Gao
- Department of Marine Organism Taxonomy and Phylogeny, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jike Lu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
26
|
MicroRNA-340 inhibits squamous cell carcinoma cell proliferation, migration and invasion by downregulating RhoA. J Dermatol Sci 2018; 92:197-206. [PMID: 30262127 DOI: 10.1016/j.jdermsci.2018.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/23/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND MicroRNAs are reported to play an important role in tumor growth and metastasis, including squamous cell carcinoma (SCC). Accumulative evidence has revealed that dysregulated miR-340 expression contributed to the carcinogenesis and development of various cancers. OBJECTIVE The aim of the current study was to investigate the role and the underlying mechanism of miR-340 in SCC cell proliferation, migration and invasion. METHODS Quantitative real-time PCR was performed to examine the expression of miR-340 in SCC tissues and cell lines. The function of miR-340 in SCC was investigated through Cell Counting Kit-8, wound healing, transwell migration and invasion assays. Bioinformatics analysis, luciferase reporter assay, western blotting and immunohistochemical analysis were conducted to predict and confirm the target gene of miR-340. RESULTS In the present study, we first found that miR-340 was significantly decreased in both SCC tissues and cell lines. Moreover, ectopic expression of miR-340 remarkably attenuated SCC cell proliferation, migration and invasion, whereas inhibition of endogenous miR-340 promoted SCC cell proliferation, migration and invasion in vitro. Our subsequent bioinformatics analysis and luciferase reporter assay showed that RhoA was a novel direct target of miR-340 in SCC cells, and the knockdown of RhoA expression rescued the effects of miR-340 inhibition on SCC cell proliferation, migration and invasion. More importantly, the expression of RhoA and miR-340 was negatively correlated in SCC tissues. CONCLUSION Our findings demonstrate the tumor suppressor role of miR-340 in SCC by directly regulating RhoA. Therefore, restoration of miR-340 expression can be a potential therapeutic approach for SCC treatment.
Collapse
|
27
|
Lu Z, Ren Y, Zhang M, Fan T, Wang Y, Zhao Q, Liu HM, Zhao W, Hou G. FLI-06 suppresses proliferation, induces apoptosis and cell cycle arrest by targeting LSD1 and Notch pathway in esophageal squamous cell carcinoma cells. Biomed Pharmacother 2018; 107:1370-1376. [PMID: 30257352 DOI: 10.1016/j.biopha.2018.08.140] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/15/2022] Open
Abstract
Aberrant activation of the Notch signaling plays an important role in progression of esophageal squamous cell carcinoma (ESCC) and may represent a potential therapeutic target for ESCC. FLI-06 is a novel Notch inhibitor, preventing the early secretion of Notch signaling. However, little information about the antitumor activity of FLI-06 has been reported so far. To evaluate the anti-tumor activity and possible molecular mechanism of FLI-06 to ESCC cells, the effects of FLI-06 on cell viability, apoptosis and cell cycle were evaluated by CCK-8 and flow cytometry assays, respectively, in ESCC cell lines ECa109 and EC9706, and the expressions of proteins in Notch signaling pathway and LSD1 were investigated after cells were treated with FLI-06 by Western blotting. The results showed that FLI-06 blocked proliferation, induced apoptosis and G1 phase arrest of ESCC cells in a dose-dependent manner. Mechanistically, we found FLI-06 could inhibit Notch signaling pathway by decreasing the expressions of Notch3, DTX1 and Hes1. Interestingly, we also found that the expression of LSD1 (histone lysine specific demethylase 1), which is dysregulated in multiple tumors, was also inhibited by FLI-06. In addition, inhibition of Notch pathway by γ-secretase inhibitor GSI-DAPT could also inhibit LSD1 expression. The current study demonstrated that FLI-06 exerts antitumor activity on ESCC by inhibiting both LSD1 and Notch pathway, which provides the theory support for the treatment of ESCC with FLI-06.
Collapse
Affiliation(s)
- Zhaoming Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of Cancer Chemoprevention, Henan Province, Zhengzhou 450001, China
| | - Yandan Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengying Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tianli Fan
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yang Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qi Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Co-Innovation Center of Henan Province for New Drug R & D and Preclinical Safety, Zhengzhou University, Zhengzhou, China
| | - Wen Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Co-Innovation Center of Henan Province for New Drug R & D and Preclinical Safety, Zhengzhou University, Zhengzhou, China
| | - Guiqin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Co-Innovation Center of Henan Province for New Drug R & D and Preclinical Safety, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
28
|
Zhao X, Xian Y, Wang C, Ding L, Meng X, Zhu W, Hang S. Calcium-sensing receptor-mediated L-tryptophan-induced secretion of cholecystokinin and glucose-dependent insulinotropic peptide in swine duodenum. J Vet Sci 2018; 19:179-187. [PMID: 29284209 PMCID: PMC5879066 DOI: 10.4142/jvs.2018.19.2.179] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/17/2017] [Accepted: 12/26/2017] [Indexed: 12/31/2022] Open
Abstract
This study aimed to elucidate the effect of tryptophan (Trp) on gut hormone secretion as well as the roles of the calcium-sensing receptor (CaSR) and its downstream signaling pathway in gut hormone secretion by assessing swine duodenal perfusion in vitro. Swine duodenum was perfused with Krebs-Henseleit buffer as a basal solution. Various concentrations (0, 10, and 20 mM) of Trp were applied to investigate its effect on gut hormone secretion. A CaSR antagonist was used to detect the involvement of CaSR and its signal molecules. The 20 mM Trp concentration promoted the secretion of cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP), elevated the mRNA level of CaSR, and upregulated the protein levels of CaSR, protein kinase C (PKC), and inositol trisphosphate receptor (IP3R). However, NPS 2143, an inhibitor of CaSR, attenuated the CCK and GIP release, reduced the mRNA level of CaSR, and decreased the protein levels of CaSR, PKC, and IP3R with 20 mM Trp perfusion. The results indicate that CCK and GIP secretion can be induced by Trp in swine duodenum in vitro, and the effect is mediated by CaSR and its downstream signal molecules PKC and IP3R.
Collapse
Affiliation(s)
- Xiuying Zhao
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yihan Xian
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Wang
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Liren Ding
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xianglong Meng
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| | - Suqin Hang
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
29
|
Jia Z, Wang M, Wang X, Wang L, Qiu L, Song L. Transcriptome sequencing reveals the involvement of reactive oxygen species in the hematopoiesis from Chinese mitten crab Eriocheir sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 82:94-103. [PMID: 29307815 DOI: 10.1016/j.dci.2017.12.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 06/07/2023]
Abstract
Reactive oxygen species (ROS) produced in vivo during various electron transfer reactions are generally kept at a certain level since they are harmful to cells. However, it can sensitize hematopoietic progenitors to differentiation, and plays a signaling role in the regulation of hematopoietic cell fate. In the present study, the transcriptomes of crab HPT and hemocytes were sequenced using the Ion Torrent Proton sequencing platform. A total of 51,229,690 single end reads were obtained from six single-end libraries, which were assembled into 31346 unireads as reference. After mapping and transcript assembling, 362 differently expressed genes were identified and 301 of them were deemed to be more abundant in HPT. GO annotation revealed that they were mostly implicated in DNA, RNA and protein synthesis, cell division, mitochondria activities and energy metabolism. The expression level of mitochondrial complexes I (mitochondrial NADH-ubiquinone oxidoreductase) which was the main natural producers of mitochondrial ROS was found to be 8.6-fold (p < 0.01) higher in HPT than that in hemocytes. In hemocytes, the proteinase genes associated with proPO activation from the 61 up-regulated genes in hemocytes were the main up-regulated genes which might be the potential markers for mature hemocytes. ROS level in HPT cells was relatively higher which was confirmed with the high expression level of mitochondria related genes identified by transcriptome sequencing. After the ROS level was depressed by N-acetyl-l-cysteine (NAC), the production of hemocytes from HPT was inhibited, and the recovery of the total hemocytes counts was delayed. These results collectively indicated that the genes in redox system were more active in HPT, and ROS could function as an important modulator in the hematopoiesis of crab and promote the production of hemocytes from HPT.
Collapse
Affiliation(s)
- Zhihao Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Xiudan Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| | - Linsheng Song
- Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
30
|
Shi K, Zhang JZ, Zhao RL, Yang L, Guo D. PSMD7 downregulation induces apoptosis and suppresses tumorigenesis of esophageal squamous cell carcinoma via the mTOR/p70S6K pathway. FEBS Open Bio 2018; 8:533-543. [PMID: 29632807 PMCID: PMC5881544 DOI: 10.1002/2211-5463.12394] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/03/2018] [Accepted: 01/12/2018] [Indexed: 12/23/2022] Open
Abstract
PSMD7, a 19S proteasome subunit, is overexpressed in most carcinoma cells. It forms a dimer with PSMD14 that functions in the removal of attached ubiquitin chain. However, there is little knowledge about the cellular mechanism of PSMD7 and its exact biological function, especially in cancer cells. In this study, we explored the role of PSMD7 in proliferation, cell cycle, apoptosis, and proteasomal proteolysis in the esophageal squamous cell carcinoma (ESCC) cell line EC9706. Our results showed that PSMD7 was highly expressed in ESCC cells. Downregulation of PSMD7 by lentivirus‐mediated shRNA led to decreased proliferation, increased cell apoptosis, and reduced proteasomal function. Notably, lower expression level of mTOR and p70S6K and suppressed activity of mTOR/p70S6K pathway were detected after PSMD7 downregulation. By contrast, increased expression of p‐mTORSer2448 and p‐p70S6KThr421/Ser424 was discovered upon PSMD7 overexpression in Het‐1A cells. Furthermore, PSMD7 downregulation contributed to decelerated tumor growth, inhibition of proteasomal function, induced cell apoptosis and attenuated activity of mTOR/p70S6K pathway in vivo. These findings suggest that PSMD7 and the mTOR/p70S6K pathway may be a promising candidate for developing therapies for ESCC.
Collapse
Affiliation(s)
- Ke Shi
- Department of Biochemistry and Molecular Biology Henan Medical College China
| | - Jin-Zhong Zhang
- Department of Biochemistry and Molecular Biology Henan Medical College China
| | - Rui-Li Zhao
- Editorial Department of Journal of Henan University of Technology Henan University of Technology Zhengzhou China.,College of Biological Engineering Henan University of Technology Zhengzhou China
| | - Liang Yang
- Department of Microbiology and Immunology and Medicine Henan Medical College China
| | - Dan Guo
- Department of Biochemistry and Molecular Biology Henan Medical College China
| |
Collapse
|
31
|
Wang X, Sun K, Wang B. Bioactive Pimarane Diterpenes from the Arctic Fungus Eutypella sp. D-1. Chem Biodivers 2018; 15. [PMID: 29168349 DOI: 10.1002/cbdv.201700501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
Two new pimarane diterpenes, libertellenone M (1) and libertellenone N (2), together with five known compounds were isolated from the culture extract of Eutypella sp. D-1 derived from high-latitude soil of the Arctic. The structures of these compounds were determined by spectroscopic data as well as experimental and calculated electronic circular dichroism (ECD) analysis. Antimicrobial and cytotoxic activities of the isolated compounds were evaluated. Compound 3 exhibited weak antibacterial activity against Escherichia coli, Bacillus subtilis, and Vibrio vulnificus, each with MIC values of 16 μg/mL. Compounds 2 and 3 showed moderate cytotoxic activity against K562 and MCF-7 cell lines with IC50 values of 7.67 and 9.57 μm, respectively.
Collapse
Affiliation(s)
- Xiaoli Wang
- Marine Science and Technology College, Zhejiang Ocean University, 1 South Haida Road, Zhoushan, 316022, P. R. China.,School of Food and Pharmacy, Zhejiang Ocean University, 1 South Haida Road, Zhoushan, 316022, P. R. China
| | - Kunlai Sun
- School of Food and Pharmacy, Zhejiang Ocean University, 1 South Haida Road, Zhoushan, 316022, P. R. China
| | - Bin Wang
- School of Food and Pharmacy, Zhejiang Ocean University, 1 South Haida Road, Zhoushan, 316022, P. R. China
| |
Collapse
|
32
|
Li S, Hou G, Wang Y, Su X, Xue L. Influence of Recombinant Human Growth Hormone (rhGH) on Proliferation of Hepatocellular Carcinoma Cells with Positive and Negative Growth Hormone Receptorsin Vitro. TUMORI JOURNAL 2018; 96:282-8. [DOI: 10.1177/030089161009600216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background Recombinant human growth hormone (rhGH) is increasingly used in the clinic because it promotes the synthesis of proteins. However, rhGH is able to increase malignant transformation and tumor recurrence. The aim of this study was to investigate the effects of rhGH on hepatocellular carcinoma (HCC) cells with positive and negative growth hormone receptors (GHR) in order to guide its clinical application. Methods and study design Cells of the human HCC cell lines Bel-7402 (GHR+) and SMMC-7721 (GHR-) as well as human umbilical vein endothelial cell line ECV304 cells in the exponential growth phase were harvested and divided into experimental and control groups. After the human HCC cells were cultured alone or co-cultured with ECV304 cells under the different treatments, cell cycle phase, proliferation index, and expression levels of vascular endothelial growth factor (VEGF) mRNA and proteins were determined. Results In the Bel-7402 GHR+ cells treated with rhGH, both the percentage of cell in G2-M phase and the proliferation index were higher than those of controls (P <0.05); this was not the case in the SMMC-7721 GHR- cells treated with rhGH (P >0.05). Although there was no difference in the cell doubling times between ECV304 cells co-incubated with Bel-7721 GHR- cells treated with rhGH and without rhGH, the doubling times of ECV304 cells co-incubated with Bel-7402 GHR+ cells, when treated with rhGH, were significantly shortened compared to those of controls (P <0.05). The cell doubling times of ECV304 cells co-incubated with Bel-7721 GHR- or Bel-7402 GHR+ cells which were treated with bevacizumab were longer than those of controls and of cells with rhGH (P <0.05). The VEGF mRNA and protein expression levels were higher in Bel-7402 GHR+ cells treated with different doses of rhGH than controls (P <0.05 or P <0.01); however, there was no statistically significant difference in the expression levels of VEGF mRNA and proteins between SMMC-7721 GHR- cells treated with rhGH and controls. Conclusions rhGH can induce VEGF secretion and stimulate proliferation of Bel-7402 GHR+ cells in vitro, but has little effect on the proliferation of SMMC-7721 GHR-cells, suggesting that rhGH may be applied safely to treatment for the catabolic state in patients with GHR-negative HCC.
Collapse
Affiliation(s)
- Suyi Li
- Institute of Tumor Molecular Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou
- Cancer Center, School of Clinical Medicine, Southeast University, Nanjing
| | - Guiqing Hou
- School of Pharmaceutical Sciences, Department of Biology, Zhengzhou University, Zhengzhou, China
| | - Ying Wang
- Cancer Center, School of Clinical Medicine, Southeast University, Nanjing
| | - Xiangyu Su
- Cancer Center, School of Clinical Medicine, Southeast University, Nanjing
| | - Lexun Xue
- Institute of Tumor Molecular Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou
- Laboratory for Cell Biology, Department of Biology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Ji YM, Zhou XF, Zhang J, Zheng X, Li SB, Wei ZQ, Liu T, Cheng DL, Liu P, Song K, Tan T, Zhu H, Guo JL. DEPTOR suppresses the progression of esophageal squamous cell carcinoma and predicts poor prognosis. Oncotarget 2017; 7:14188-98. [PMID: 26893358 PMCID: PMC4924707 DOI: 10.18632/oncotarget.7420] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/29/2016] [Indexed: 01/14/2023] Open
Abstract
As a naturally occurring inhibitor of mTOR, accumulated evidence has suggested that DEPTOR plays a pivotal role in suppressing the progression of human malignances. However, the function of DEPTOR in the development of esophageal squamous cell carcinoma (ESCC) is still unclear. Here we report that the expression of DEPTOR is significantly reduced in tumor tissues derived from human patients with ESCC, and the downregulation of DEPTOR predicts a poor prognosis of ESCC patients. In addition, we found that the expression of DEPTOR negatively regulates the tumorigenic activities of ESCC cell lines (KYSE150, KYSE510 and KYSE190). Furthermore, ectopic DEPTOR expression caused a significant suppression of the cellular proliferation, migration and invasion of KYSE150 cells, which has the lowest expression level of DEPTOR in the three cell lines. Meanwhile, CRISPR/Cas9 mediated knockout of DEPTOR in KYSE-510 cells significantly promoted cellular proliferation, migration and invasion. In addition, in vivo assays further revealed that tumor growth was significantly inhibited in xenografts with ectopic DEPTOR expression as compared to untreated KYSE150 cells, and was markedly enhanced in DEPTOR knockout KYSE-510 cells. Biochemical studies revealed that overexpression of DEPTOR led to the suppression of AKT/mTOR pathway as evidenced by reduced phosphorylation of AKT, mTOR and downstream SGK1, indicating DEPTOR might control the progression of ESCC through AKT/mTOR signaling pathway. Thus, these findings, for the first time, demonstrated that DEPTOR inhibits the tumorigenesis of ESCC cells and might serve as a potential therapeutic target or prognostic marker for human patients with ESCC.
Collapse
Affiliation(s)
- Yan-Mei Ji
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Xue-Feng Zhou
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jun Zhang
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Xiang Zheng
- Department of Intensive Care Unit, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Sheng-Bao Li
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Zhi-Qiang Wei
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Tao Liu
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Dong-Liang Cheng
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Ping Liu
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Kuncheng Song
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jia-Long Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, People's Republic of China
| |
Collapse
|
34
|
The aldehyde group of gossypol induces mitochondrial apoptosis via ROS-SIRT1-p53-PUMA pathway in male germline stem cell. Oncotarget 2017; 8:100128-100140. [PMID: 29245965 PMCID: PMC5725007 DOI: 10.18632/oncotarget.22044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/05/2017] [Indexed: 01/14/2023] Open
Abstract
As a widely grown economic crop, cotton is the major oil and protein resource for human and livestock. But the highly toxic of gossypol in cottonseed severely restricts its effective utilization, consequently creating huge resource waste. Previous studies have shown the male germline stem cells were the most vulnerable cells in gossypol damages, but the mechanism was still unclear. We found gossypol induced cell viability decline resulted from apoptosis. And the increase of Caspase-9 activity in gossypol treatment hinted the mitochondrial apoptosis. So the mitochondrial dysfunction was confirmed by the decreased mitochondrial membrane potential and ATP concentration. We found the higher intracellular H2O2 level did not accompany with the O2·- associated increase in gossypol-treated, which indicated that gossypol obstructed the intracellular reactive oxygen species (ROS) elimination. Manipulated gossypol-induced H2O2 level by H2O2 and α-lipoic acid, we demonstrated that the mitochondrial dysfunction resulted from the excessive intracellular H2O2. Treated with Apogossypolone (ApoG2), an aldehyde group removed derivative of gossypol, the GSH/GSSG ratio and H2O2 did not decrease. ApoG2 also did not cause the mitochondrial apoptosis. So the aldehyde group is key factor in gossypol cytotoxicity. We respectively detected the NAD+/NADH ratio, SIRT1 activity, the relative protein level and apoptosis. Comparing with the specific inhibitors groups, the data illustrated that gossypol induced apoptosis through SIRT1-P53-PUMA pathway. This study helped to overcome barriers of gossypol cytotoxicity, which is crucial in feed and food use of cottonseed. This also provides a reference for the gossypol derivatives using in male contraception and anticancer.
Collapse
|
35
|
Luteolin, a natural flavonoid, inhibits methylglyoxal induced apoptosis via the mTOR/4E-BP1 signaling pathway. Sci Rep 2017; 7:7877. [PMID: 28801605 PMCID: PMC5554232 DOI: 10.1038/s41598-017-08204-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 07/10/2017] [Indexed: 01/28/2023] Open
Abstract
Methylglyoxal (MG) accumulation has been observed in human cerebrospinal fluid and body tissues under hyperglycaemic conditions. Recent research has demonstrated that MG-induces neuronal cell apoptosis, which promotes the development of diabetic encephalopathy. Our previous animal study has shown that luteolin, a natural flavonoid, attenuates diabetes-associated cognitive dysfunction. To further explore the neuroprotective properties of luteolin, we investigated the inhibitive effect of luteolin on MG-induced apoptosis in PC12 neuronal cells. We found that MG inhibited cell viability in a dose-dependent manner and induced apoptosis in PC12 cells. Pretreatment with Luteolin significantly elevated cell viability, reduced MG-induced apoptosis, inhibited the activation of the mTOR/4E-BP1 signaling pathway, and decreased pro-apoptotic proteins, Bax, Cytochrome C as well as caspase-3. Furthermore, we found that pretreatment with the mTOR inhibitor, rapamycin, significantly reduced the expression of the pro-apoptotic protein Bax. Therefore, these observations unambiguously suggest that the inhibitive effect of Luteolin against MG-induced apoptosis in PC12 cells is associated with inhibition of the mTOR/4E-BP1 signaling pathway.
Collapse
|
36
|
Li Y, Song X, Wang W, Wang L, Yi Q, Jiang S, Jia Z, Du X, Qiu L, Song L. The hematopoiesis in gill and its role in the immune response of Pacific oyster Crassostrea gigas against secondary challenge with Vibrio splendidus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 71:59-69. [PMID: 28159592 DOI: 10.1016/j.dci.2017.01.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 06/06/2023]
Abstract
Increasing evidences have demonstrated that the invertebrate gill is a predominant tissue participating in the immune response during pathogen challenge. In the present study, the hematopoiesis and immune activities in gill of Pacific oyster Crassostrea gigas were investigated. Stem-like cells with big nuclei and thin cytoplasm were found in the tubules of gill filaments, where DNA synthesis is active and hemocytes production are exuberant. The oysters primarily stimulated by formaldehyde-killed Vibrio splendidus exhibited stronger immune responses and enhanced cell regeneration in gill when they encountered the secondary challenge of live V. splendidus. After the secondary stimulation with V. splendidus, the expression levels of CgClec-4 and CgIFN in the gill of oysters pre-stimulated with formaldehyde-killed V. splendidus were significantly higher (p < 0.05) than that in the oysters pre-stimulated with filter-sterilized (0.22 μm pore size) sea water, while the expression level of CgIL-17 was significantly decreased (p < 0.05). Meanwhile, the protein expression level of hematopoietic transcription factor CgGATA3 and immune-related protein CgEcSOD in gill increased apparently after the secondary challenge with V. splendidus. ROS production was also enhanced (p < 0.05) at 6 h and 24 h after the secondary challenge. The phagocytic rate in gill of oysters pre-stimulated with formaldehyde-killed V. splendidus was significantly increased (p < 0.05) at 6 h after the secondary challenge with live V. splendidus, showing faster response than that pre-stimulated with filter-sterilized sea water. These results collectively showed that the immune parameters in gill were apparently enhanced after secondary challenge with live V. splendidus, indicating that hematopoiesis might participate in immune priming in Pacific oyster C. gigas.
Collapse
Affiliation(s)
- Yiqun Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaorui Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China.
| | - Qilin Yi
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Shuai Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Zhihao Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyu Du
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
37
|
OP16, a novel ent-kaurene diterpenoid, potentiates the antitumor effect of rapamycin by inhibiting rapamycin-induced feedback activation of Akt signaling in esophageal squamous cell carcinoma. Biochem Pharmacol 2017; 140:16-27. [PMID: 28539264 DOI: 10.1016/j.bcp.2017.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/17/2017] [Indexed: 11/22/2022]
Abstract
Hyperactivation of mTOR signaling pathway has been viewed as a significant molecular pathogenesis of cancer. However, inhibition of mTOR by rapamycin and its analogs could induce numerous negative feedback loops to attenuate their therapeutic efficacy. As a traditional Chinese herbal medicine, Rabdosia rubescens has been used to treat esophageal squamous cell carcinoma (ESCC) for hundreds of years, and its major effective component is oridonin. Here we reported that OP16, a novel analog of oridonin, showed potent inhibition of cell proliferation and Akt phosphorylation in ESCC cells. The combination of OP16 and rapamycin possesses synergistic anti-proliferative and pro-apoptotic effects both in ESCC cells and ESCC xenografts, and no obvious adverse effect was observed in vivo. Mechanistic analysis revealed that OP16 could inhibit rapamycin-induced Akt activation through the p70S6K-mediated negative feedback loops, and the combination of OP16 and rapamycin was more effective in activating caspase-dependent apoptotic signaling cascade. This study supports the combined use of OP16 with rapamycin as a feasible and effective therapeutic approach for future treatment of ESCC.
Collapse
|
38
|
miR-195 Regulates Proliferation and Apoptosis through Inhibiting the mTOR/p70s6k Signaling Pathway by Targeting HMGA2 in Esophageal Carcinoma Cells. DISEASE MARKERS 2017; 2017:8317913. [PMID: 28487599 PMCID: PMC5402242 DOI: 10.1155/2017/8317913] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/13/2017] [Accepted: 03/13/2017] [Indexed: 12/19/2022]
Abstract
miR-195 is related to tumorigenesis and frequently inhibits cell proliferation and promotes apoptosis in various cancers, including esophageal carcinoma (EC). The mTOR/p70s6k signaling pathway, which is the major target pathway for HMGA2, regulates the survival and cell proliferation of many tumors and is commonly active in EC. The relationships of miR-195, HMGA2, and the mTOR/p70s6k signaling pathway in EC, however, remain unknown. In the present study, we found that the miR-195 level was significantly downregulated in EC tissues, while the mRNA expressions of HMGA2 were significantly upregulated. Dual-luciferase reporter assay demonstrated that HMGA2 is a target of miR-195. MTT assay and flow cytometry revealed that miR-195 overexpression inhibited cell proliferation and induced apoptosis by targeting HMGA2. We also found that HMGA2 restored the inhibitory effect of miR-195 on phosphorylation of mTOR and p70S6K. Furthermore, rapamycin, a specific inhibitor of the mTOR/p70S6K signaling pathway, decreased the levels of Ki-67 and Bcl-2/Bax ratio, inhibited cell proliferation, and promoted apoptosis in EC cells. In conclusion, upregulation of miR-195 significantly suppressed cell growth and induced apoptosis of EC cells via suppressing the mTOR/p70s6k signaling pathway by targeting HMGA2.
Collapse
|
39
|
Tang ZM, Zhai XX, Ding JC. Expression of mTOR/70S6K signaling pathway in pathological scar fibroblasts and the effects of resveratrol intervention. Mol Med Rep 2017; 15:2546-2550. [PMID: 28447760 PMCID: PMC5428871 DOI: 10.3892/mmr.2017.6339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022] Open
Abstract
The aim of the study was to examine the expression of mammalian target of rapamycin (mTOR)/70S6K signaling pathway in pathological scar fibroblasts and the effects of resveratrol (Res) intervention. The mTOR and 70S6K in pathological scar and normal skin fibroblasts were detected by immunofluorescence following treatment with different concentrations of Res. RT-PCR and western blot analysis were used to detect the expression of mTOR and 70S6K mRNA and protein, respectively. Immunofluorescence showed that the expression of 70S6K and mTOR was significantly enhanced in pathological scar fibroblasts, and mainly expressed in the nucleus, but not in normal skin fibroblasts. RT-PCR and western blot analysis showed that after different concentrations of Res treatments, the mTOR and 70S6K mRNA and protein expression significantly (P<0.05) decreased in a dose-dependent manner. In conclusion, the expression of mTOR/70S6K signaling pathway in pathological scar fibroblasts was significantly enhanced. Res can downregulate the expression of mTOR and 70S6K to achieve the inhibition of pathological scar fibroblast proliferation.
Collapse
Affiliation(s)
- Zhi-Ming Tang
- Department of Dermatology, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221003, P.R. China
| | - Xiao-Xiang Zhai
- Department of Dermatology, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221003, P.R. China
| | - Ji-Cun Ding
- Department of Burns and Plastic Surgery, The Affiliated Xuzhou Center Hospital of Nanjing University of Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
40
|
Xu F, Wang Y, Tao T, Song D, Liu X. Calreticulin attenuated microwave radiation-induced human microvascular endothelial cell injury through promoting actin acetylation and polymerization. Cell Stress Chaperones 2017; 22:87-97. [PMID: 27815707 PMCID: PMC5225063 DOI: 10.1007/s12192-016-0745-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 11/26/2022] Open
Abstract
Recent work reveals that actin acetylation modification has been linked to different normal and disease processes and the effects associated with metabolic and environmental stressors. Herein, we highlight the effects of calreticulin on actin acetylation and cell injury induced by microwave radiation in human microvascular endothelial cell (HMEC). HMEC injury was induced by high-power microwave of different power density (10, 30, 60, 100 mW/cm2, for 6 min) with or without exogenous recombinant calreticulin. The cell injury was assessed by lactate dehydrogenase (LDH) activity and Cell Counting Kit-8 in culture medium, migration ability, intercellular junction, and cytoskeleton staining in HMEC. Western blotting analysis was used to detected calreticulin expression in cytosol and nucleus and acetylation of globular actin (G-actin). We found that HMEC injury was induced by microwave radiation in a dose-dependent manner. Pretreatment HMEC with calreticulin suppressed microwave radiation-induced LDH leakage and increased cell viability and improved microwave radiation-induced decrease in migration, intercellular junction, and cytoskeleton. Meanwhile, pretreatment HMEC with exogenous calreticulin upregulated the histone acetyltransferase activity and the acetylation level of G-actin and increased the fibrous actin (F-actin)/G-actin ratio. We conclude that exogenous calreticulin protects HMEC against microwave radiation-induced injury through promoting actin acetylation and polymerization.
Collapse
Affiliation(s)
- Feifei Xu
- Department of Pathophysiology, Chinese PLA General Hospital, Beijing, China
| | - You Wang
- Department of Pathophysiology, Chinese PLA General Hospital, Beijing, China
| | - Tianqi Tao
- Department of Pathophysiology, Chinese PLA General Hospital, Beijing, China
| | - Dandan Song
- Department of Pathophysiology, Chinese PLA General Hospital, Beijing, China
| | - Xiuhua Liu
- Department of Pathophysiology, Chinese PLA General Hospital, Beijing, China.
- State Key Laboratory of Kidney Disease, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
41
|
Li Q, Yang L, Han K, Zhu L, Zhang Y, Ma S, Zhang K, Yang B, Guan F. Ets2 knockdown inhibits tumorigenesis in esophageal squamous cell carcinoma in vivo and in vitro. Oncotarget 2016; 7:61458-61468. [PMID: 27556183 PMCID: PMC5308664 DOI: 10.18632/oncotarget.11369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 08/08/2016] [Indexed: 01/13/2023] Open
Abstract
Increased expression of Ets2 is reported upregulated in esophageal squamous cell carcinoma tissue. However, the function of Ets2 in carcinogenesis of ESCC is poorly understood. Here, the rise of Ets2 was confirmed in ESCC cells and Ets2 depletion by RNA interference promotes cell apoptosis, inhibits cell proliferation, attenuates cell invasion and induces cell cycle G0/G1 arrest in vitro. Moreover, in vivo, Xenograft mouse model studies showed Ets2 knockdown inhibits tumor formation and metastasis significantly. Furthermore, Ets2 depletion inactivates the mTOR/p70S6K signaling pathway both in vitro and in vivo. Taken together, these findings strongly suggest that a critical role of Ets2 in human ESCC pathogenesis via the inactivation of the mTOR/p70S6K signaling pathway.
Collapse
Affiliation(s)
- Qinghua Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Lu Yang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Kang Han
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Liqiang Zhu
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan Province, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Kun Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Fangxia Guan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.,School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
42
|
Zhang Y, Zhu X, Huang T, Chen L, Liu Y, Li Q, Song J, Ma S, Zhang K, Yang B, Guan F. β-Carotene synergistically enhances the anti-tumor effect of 5-fluorouracil on esophageal squamous cell carcinoma in vivo and in vitro. Toxicol Lett 2016; 261:49-58. [PMID: 27586268 DOI: 10.1016/j.toxlet.2016.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/08/2016] [Accepted: 08/14/2016] [Indexed: 12/26/2022]
Abstract
Recently, we reported that β-carotene exhibited anticancer activity against human esophageal squamous cell carcinoma cells in vitro. In the present study, we examined a novel therapeutic strategy by combining β-carotene with 5-fluorouracil (5-FU) in human esophageal cancer in vitro and in vivo, and elucidated the underlying mechanisms. We found that the combination of 5-FU and β-carotene displayed greater growth inhibitory effects than did either compound alone in esophageal squamous cell carcinoma (ESCC) cells. In addition, the combination of 5-FU and β-carotene displayed greater tumor growth inhibition in an Eca109 xenograft mouse model than did a single agent with low systemic toxicity. β-Carotene enhanced 5-FU-induced apoptosis. TUNEL staining revealed that the rate of TUNEL-positive cells was markedly increased in tumor tissues after treatment with 5-FU and β-carotene. Western blotting and immunohistochemistry revealed the down-regulation of Bcl-2 and PCNA and the up-regulation of Bax and caspase-3 in tumor tissues. Further studies demonstrated that the combined administration of 5-FU and β-carotene significantly down-regulated the protein levels of Cav-1, p-AKT, p-NF-κB, p-mTOR and p-p70S6K in Eca109 cells more effectively than did 5-FU alone. These data suggested that the combined therapy of 5-FU and β-carotene exerted synergistic antitumor effects in vivo and in vitro and could constitute a novel therapeutic treatment for ESCC.
Collapse
Affiliation(s)
- Yanting Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xiangzhan Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Tuanjie Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Lei Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yanxia Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Qinghua Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jishi Song
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Kun Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Fangxia Guan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
43
|
Zuo Y, Yu X, Lu S. Dermal Fibroblasts from Different Layers of Pig Skin Exhibit Different Profibrotic and Morphological Characteristics. Anat Rec (Hoboken) 2016; 299:1585-1599. [PMID: 27518880 DOI: 10.1002/ar.23458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 02/04/2023]
Abstract
In vitro studies of human dermal fibroblast (DF) heterogeneity have long been reported, yet in vivo studies and related research on animals are rare. The objectives of the study were to determine whether the DFs of pigs exhibit heterogeneity and to identify an animal model for the in vivo study of DF heterogeneity. The skin of three female red Duroc pigs (FRDPs) was separated into six layers, and the second and fifth layers (i.e., the superficial and deep dermis) were used in the establishment of wound models and cell cultures. To create the wound models, 54 tongue-shaped flaps were created on one side of the dorsum, and the underlying dermis was then fully replaced with the superficial or deep dermis (the superficial and deep groups, respectively). Skin samples were harvested at postoperative weeks 1, 2, and 3 for measurements of the normal and wounded skin thicknesses. Cells cultured from the superficial and deep dermis (i.e., superficial and deep DFs) were subjected to quantitative estimation of collagen and electron microscopy. The wounded skin thickness in the deep group was significantly greater than that in the superficial group. In contrast with the long deep DFs, the superficial DFs were short and exhibited microvilli-like cell surface projections. Compared with the superficial DFs, the deep DFs exhibited a greater density of rough endoplasmic reticulum and produced significantly more collagen. Similar to humans, FRDPs exhibit DF heterogeneity and should thus be a good animal model for in vivo studies of DF heterogeneity. Anat Rec, 299:1585-1599, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yanhai Zuo
- Shanghai Burns Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
| | - Xiaoping Yu
- Shanghai Burns Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China
| | - Shuliang Lu
- Shanghai Burns Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
44
|
Downregulation of p70S6K Enhances Cell Sensitivity to Rapamycin in Esophageal Squamous Cell Carcinoma. J Immunol Res 2016; 2016:7828916. [PMID: 27595116 PMCID: PMC4993948 DOI: 10.1155/2016/7828916] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
It has been demonstrated that mTOR/p70S6K pathway was abnormally activated in many cancers and rapamycin and its analogs can restrain tumor growth through inhibiting this pathway, but some tumors including esophageal squamous cell carcinoma (ESCC) appear to be insensitive to rapamycin in recent studies. In the present study, we explored the measures to improve the sensitivity of ESCC cells to rapamycin and identified the clinical significance of the expression of phosphorylated p70S6K (p-p70S6K). The results showed that, after downregulating the expression of p70S6K and p-p70S6K by p70S6K siRNA, the inhibitory effects of rapamycin on cell proliferation, cell cycle, and tumor growth were significantly enhanced in vitro and in vivo. Furthermore, p-p70S6K had strong positive expression in ESCC tissues and its expression was closely related to lymph node metastasis and the TNM staging. These results indicated that p-p70S6K may participate in the invasion and metastasis in the development of ESCC and downregulation of the expression of p-p70S6K could improve the sensitivity of cells to rapamycin in ESCC.
Collapse
|
45
|
Wang YW, Zhang JH, Yu Y, Yu J, Huang L. Inhibition of Store-Operated Calcium Entry Protects Endothelial Progenitor Cells from H2O2-Induced Apoptosis. Biomol Ther (Seoul) 2016; 24:371-9. [PMID: 27169819 PMCID: PMC4930280 DOI: 10.4062/biomolther.2015.130] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/09/2015] [Accepted: 11/17/2015] [Indexed: 01/09/2023] Open
Abstract
Store-operated calcium entry (SOCE), a major mode of extracellular calcium entry, plays roles in a variety of cell activities. Accumulating evidence indicates that the intracellular calcium ion concentration and calcium signaling are critical for the responses induced by oxidative stress. The present study was designed to investigate the potential effect of SOCE inhibition on H2O2-induced apoptosis in endothelial progenitor cells (EPCs), which are the predominant cells involved in endothelial repair. The results showed that H2O2-induced EPC apoptosis was reversed by SOCE inhibition induced either using the SOCE antagonist ML-9 or via silencing of stromal interaction molecule 1 (STIM1), a component of SOCE. Furthermore, SOCE inhibition repressed the increases in intracellular reactive oxygen species (ROS) levels and endoplasmic reticulum (ER) stress and ameliorated the mitochondrial dysfunction caused by H2O2. Our findings provide evidence that SOCE inhibition exerts a protective effect on EPCs in response to oxidative stress induced by H2O2 and may serve as a potential therapeutic strategy against vascular endothelial injury.
Collapse
Affiliation(s)
- Yan-Wei Wang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Ji-Hang Zhang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Yang Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Jie Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Lan Huang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| |
Collapse
|
46
|
Zhu J, Wang M, He J, Zhu M, Wang JC, Jin L, Wang XF, Yang YJ, Xiang JQ, Wei Q. Polymorphisms in the AKT1 and AKT2 genes and oesophageal squamous cell carcinoma risk in an Eastern Chinese population. J Cell Mol Med 2016; 20:666-77. [PMID: 26828791 PMCID: PMC5126231 DOI: 10.1111/jcmm.12750] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/02/2015] [Indexed: 02/05/2023] Open
Abstract
Ethnic Han Chinese are at high risk of developing oesophageal squamous cell carcinoma (ESCC). Aberrant activation of the AKT signalling pathway is involved in many cancers, including ESCC. Some single nucleotide polymorphisms (SNPs) in genes involved in this pathway may contribute to ESCC susceptibility. We selected five potentially functional SNPs in AKT1 (rs2494750, rs2494752 and rs10138277) and AKT2 (rs7254617 and rs2304186) genes and investigated their associations with ESCC risk in 1117 ESCC cases and 1096 controls in an Eastern Chinese population. None of individual SNPs exhibited an association with ESCC risk. However, the combined analysis of three AKT1 SNPs suggested that individuals carrying one of AKT1 variant genotypes had a decreased ESCC risk [adjusted odds ratio (OR) = 0.60, 95% CI = 0.42-0.87]. Further stratified analysis found that AKT1 rs2294750 SNP was associated with significantly decreased ESCC risk among women (adjusted OR = 0.63, 95% CI = 0.43-0.94) and non-drinkers (OR = 0.79, 95% CI = 0.64-0.99). Similar protective effects on women (adjusted OR = 0.56, 95% CI = 0.37-0.83) and non-drinker (adjusted OR = 0.75, 95% CI = 0.60-0.94) were also observed for the combined genotypes of AKT1 SNPs. Consistently, logistic regression analysis indicated significant gene-gene interactions among three AKT1 SNPs (P < 0.015). A three-AKT1 SNP haplotype (C-A-C) showed a significant association with a decreased ESCC risk (adjusted OR = 0.70, 95% CI = 0.52-0.94). Multifactor dimensionality reduction analysis confirmed a high-order gene-environment interaction in ESCC risk. Overall, we found that three AKT1 SNPs might confer protection against ESCC risk; nevertheless, these effects may be dependent on other risk factors. Our results provided evidence of important gene-environment interplay in ESCC carcinogenesis.
Collapse
Affiliation(s)
- Jinhong Zhu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Molecular Epidemiology Laboratory and Department of Laboratory Medicine, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Mengyun Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jiu-Cun Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Li Jin
- Ministry of Education Key Laboratory of Contemporary Anthropology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xiao-Feng Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Ya-Jun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Jia-Qing Xiang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
47
|
Li Y, Jiang S, Li M, Xin L, Wang L, Wang H, Qiu L, Song L. A cytokine-like factor astakine accelerates the hemocyte production in Pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 55:179-187. [PMID: 26523496 DOI: 10.1016/j.dci.2015.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 06/05/2023]
Abstract
Astakine has been reported to be a hematopoietic growth factor of prokineticin homolog firstly found in arthropods freshwater crayfish Pacifastacus leniusculus. In the present study, an astakine homologous gene was identified from Pacific oyster Crassostrea gigas (designated CgAstakine). The full length cDNA of CgAstakine encoded a polypeptide of 103 amino acids containing a prokineticin (PK) domain homologous to that in astakine from freshwater crayfish P. leniusculus. The deduced amino acid sequence of CgAstakine shared higher similarity with those of other invertebrate astakines than prokineticins from vertebrates. The mRNA of CgAstakine was highly expressed in hepatopancreas and adductor muscle of oyster, while the CgAstakine protein was mainly distributed in hepatopancreas, gill and hemocytes. The mRNA expression of CgAstakine in hemocytes was significantly increased (p < 0.01) and maintained at a high level from 3 h to 9 h after Vibrio anguillarum challenge. After the oyster hemocytes were incubated with 5 μg/mL recombinant CgAstakine protein (rCgAstakine) for 24 h in vitro, the proliferation of hemocytes was significantly increased to 1.89 fold of that in control group (p < 0.05). Moreover, the total count of oyster hemocytes was significantly upregulated (2.45 fold of that in control group, p < 0.05) at 12 h after the oysters were received an injection of rCgAstakine (0.5 μg/g). These results collectively indicated that CgAstakine could modulate the hemocytes proliferation both in vitro and in vivo, and probably involved in the hematopoietic process fighting against the invasion of foreign pathogens.
Collapse
Affiliation(s)
- Yiqun Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Meijia Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lusheng Xin
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingling Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| | - Hao Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Linsheng Song
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
48
|
mTOR/P70S6K promotes spermatogonia proliferation and spermatogenesis in Sprague Dawley rats. Reprod Biomed Online 2016; 32:207-17. [DOI: 10.1016/j.rbmo.2015.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/07/2015] [Accepted: 11/10/2015] [Indexed: 01/14/2023]
|
49
|
Chen XG, Lv YX, Zhao D, Zhang L, Zheng F, Yang JY, Li XL, Wang L, Guo LY, Pan YM, Yan YW, Chen SY, Wang JN, Tang JM, Wan Y. Vascular endothelial growth factor-C protects heart from ischemia/reperfusion injury by inhibiting cardiomyocyte apoptosis. Mol Cell Biochem 2016; 413:9-23. [DOI: 10.1007/s11010-015-2622-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/08/2015] [Indexed: 11/29/2022]
|
50
|
Dai M, Feng M, Ye Y, Wu X, Liu D, Liao M, Cao W. Exogenous avian leukosis virus-induced activation of the ERK/AP1 pathway is required for virus replication and correlates with virus-induced tumorigenesis. Sci Rep 2016; 6:19226. [PMID: 26754177 PMCID: PMC4709637 DOI: 10.1038/srep19226] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/03/2015] [Indexed: 12/13/2022] Open
Abstract
A proteomics approach was used to reveal the up-regulated proteins involved in the targeted mitogen-activated protein kinase (MAPK) signal transduction pathway in DF-1 cells after ALV subgroup J (ALV-J) infection. Next, we found that ALV-J CHN06 strain infection of DF-1 cells correlated with extracellular signal-regulated kinase 2 (ERK2) activation, which was mainly induced within 15 min, a very early stage of infection, and at a late infection stage, from 108 h to 132 h post-infection. Infection with other ALV subgroup (A/B) strains also triggered ERK/MAPK activation. Moreover, when activating ERK2, ALV subgroups A, B and J simultaneously induced the phosphorylation of c-Jun, an AP1 family member and p38 activation but had no obvious effect on JNK activation at either 15 min or 120 h. Interestingly, only PD98059 inhibited the ALV-induced c-Jun phosphorylation while SP600125 or SB203580 had no influence on c-Jun activation. Furthermore, the viral gp85 and gag proteins were found to contribute to ERK2/AP1 activation. Additionally, the specific ERK inhibitor, PD980509, significantly suppressed ALV replication, as evidenced by extremely low levels of ALV promoter activity and ALV-J protein expression. In vivo analysis of ERK2 activation in tumor cells derived from ALV-J-infected chicken demonstrated a strong correlation between ERK/MAPK activation and virus-associated tumorigenesis.
Collapse
Affiliation(s)
- Manman Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
| | - Min Feng
- College of Animal Science, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
| | - Yu Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
| | - Xiaochan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
| | - Di Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture
- South China Collaborative innovation Center for Prevention and Control of poultry Infectious diseases and Safety of Poultry Products.
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People’s Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture
- South China Collaborative innovation Center for Prevention and Control of poultry Infectious diseases and Safety of Poultry Products.
| |
Collapse
|