1
|
Gao YP, Lu JT, Zhang HJ, Cui ZM, Guo Y, Zhang X, Wang W, Qiu LL, Wang XY, Wang TY, Jia YL. MAT2A Knockdown Enhances Recombinant Protein Expression in Transgenic CHO Cells Through Regulation of Cell Cycle. Biotechnol Bioeng 2025; 122:1461-1471. [PMID: 40011400 DOI: 10.1002/bit.28962] [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: 12/14/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Chinese hamster ovary (CHO) cells represent the most widely utilized host system for industrial production of high-quality recombinant protein therapeutics. Novel CHO cell line development is achieved through genetic and cellular engineering approaches, effectively addressing limitations such as clonal variation and productivity loss during culture. Previous studies have established that MAT2A inhibition in tumor cells promotes expression of the cyclin-dependent kinase inhibitor p21, inducing antitumor activity. Notably, p21 induction has been shown to enhance recombinant protein expression in CHO cells by triggering cell cycle arrest. In this study, we identified MAT2A as a potential regulatory target, showing significant differential expression in transfected CHO cells with elevated versus diminished recombinant protein production. To investigate this phenomenon, we generated CHO cells with MAT2A knockdown (shMAT2A) and evaluated their recombinant protein output. Results demonstrated that MAT2A silencing enhanced recombiant protein/antibody production by 1.73-/1.70-fold through suppression of CyclinD1, thereby activating p21 and inducing G1 phase arrest. Furthermore, pharmacological inhibition of MAT2A using small molecules increased cell volume, boosted metabolic activity, and improved specific antibody productivity of recombiant protein/antibody production by 1.88-/2.16-fold in transfected CHO cells. These findings advance our understanding of MAT2A-mediated regulatory mechanisms and provide a strategic framework for developing high-efficiency CHO cell expression systems.
Collapse
Affiliation(s)
- Yan-Ping Gao
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Jiang-Tao Lu
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Hui-Jie Zhang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Zhao-Ming Cui
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yang Guo
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Xi Zhang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Wen Wang
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| | - Le-Le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Yin Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Yan-Long Jia
- School of Pharmacy, XinXiang Medical University, Xinxiang, China
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang, China
- Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Lei Y, Zhong C, Zhang J, Zheng Q, Xu Y, Li Z, Huang C, Ren T. Senescent lung fibroblasts in idiopathic pulmonary fibrosis facilitate non-small cell lung cancer progression by secreting exosomal MMP1. Oncogene 2025; 44:769-781. [PMID: 39663393 PMCID: PMC11888990 DOI: 10.1038/s41388-024-03236-5] [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: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Lung cancer is a fatal complication of idiopathic pulmonary fibrosis (IPF) with a poor prognosis. Current treatments are insufficient in improving the prognosis of lung cancer patients with comorbid idiopathic pulmonary fibrosis (IPF-LC). Senescent fibroblasts, as stromal cells in the tumor microenvironment, influence tumor progression via exosomes. With evidence that fibroblast senescence is an important mechanism of IPF, we investigated the impact of senescent IPF lung fibroblast (diseased human lung fibroblasts, DHLF)-derived exosomes on non-small cell lung cancer (NSCLC). We found DHLF expressed significant senescence markers, and promoted NSCLC proliferation, invasion, and epithelial-mesenchymal transition. Specifically, senescent DHLF showed strong secretion of exosomes, and these exosomes enhanced the proliferation and colony-forming ability of cancer cells. Proteomic analysis showed DHLF-derived exosomes exhibited upregulated senescence-associated secretory phenotype (SASP) factors, notably MMP1, which activates the surface receptor PAR1. Knocking down MMP1 or using PAR1 inhibitors reduced the tumor-promoting effects of DHLF-derived exosomes in vivo and in vitro. Mechanistically, MMP1 acted by activating the PI3K-AKT-mTOR pathway. In conclusion, our results suggest that exosomal MMP1 derived from senescent IPF fibroblasts promotes NSCLC proliferation and colony formation by targeting PAR1 and activating the PI3K-AKT-mTOR pathway. These findings provide a novel therapeutic approach for patients with IPF-LC.
Collapse
Affiliation(s)
- Yuqiong Lei
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Cheng Zhong
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jingyuan Zhang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qi Zheng
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yongle Xu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhoubin Li
- Department of Lung Transplantation and Thoracic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Chenwen Huang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Department of Clinical Research Centre, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Stem Cell Center, Shanghai Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
3
|
Joshi G, Yadav UP, Rafiq Z, Grewal P, Kumar M, Singh T, Jha V, Sharma P, Eriksson LA, Srinivas L, Dahibhate NL, Srivastava P, Bhutani P, Mishra UK, Sharon A, Banerjee UC, Sharma N, Chatterjee J, Tikoo K, Singh S, Kumar R. Design and Synthesis of Topoisomerases-Histone Deacetylase Dual Targeted Quinoline-Bridged Hydroxamates as Anticancer Agents. J Med Chem 2025; 68:2849-2868. [PMID: 39808731 DOI: 10.1021/acs.jmedchem.4c02135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The multifactorial nature of cancer requires treatment that involves simultaneous targeting of associated overexpressed proteins and cell signaling pathways, possibly leading to synergistic effects. Herein, we present a systematic study that involves the simultaneous inhibition of human topoisomerases (hTopos) and histone deacetylases (HDACs) by multitargeted quinoline-bridged hydroxamic acid derivatives. These compounds were rationally designed considering pharmacophoric features and catalytic sites of the cross-talk proteins, synthesized, and assessed for their anticancer potential. Our findings revealed that the compound 5c significantly produced anticancer effects in vitro and in vivo by reducing the tumor growth and its size in the A549 cell-induced lung cancer xenograft model through multiple mechanisms, primarily by multi-inhibition of hTopoI/II and HDACs, especially HDAC1 via atypical binding. The present paper discusses detailed mechanistic biological investigations, structure-activity effects supported by computational docking studies, and DMPK studies and provides future scope for lead optimization and modification.
Collapse
Affiliation(s)
- Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda 151 401, India
| | - Umesh Prasad Yadav
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Zahid Rafiq
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar 160062, India
| | - Preeti Grewal
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, S.A.S. Nagar 160062, India
| | - Manvendra Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda 151 401, India
| | - Tashvinder Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Vibhu Jha
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg 405 30, Sweden
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford BD7 1DP, U.K
| | - Praveen Sharma
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Leif A Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg 405 30, Sweden
| | | | | | | | | | - Uttam Kumar Mishra
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Ashoke Sharon
- Department of Chemistry, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Uttam C Banerjee
- Department of Pharmaceutical Technology (Biotechnology), National Institute of Pharmaceutical Education and Research, S.A.S. Nagar 160062, India
| | - Nisha Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar 160062, India
| | - Joydeep Chatterjee
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda 151 401, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar 160062, India
| | - Sandeep Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda 151 401, India
| |
Collapse
|
4
|
Ju Y, Xiao W, Mathis BJ, Shi Y. KLF4: a multifunctional nexus connecting tumor progression and immune regulation. Front Immunol 2025; 16:1514780. [PMID: 39995670 PMCID: PMC11848521 DOI: 10.3389/fimmu.2025.1514780] [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: 10/21/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Krüppel-like factors (KLFs) regulate various biological processes such as cell proliferation, migration, invasion, and differentiation as gene transcription factors. Signaling pathways which mediated by KLF4 and KLF4 have a sophisticated role in tumors due to multiple factors, including the types or stage of tumors. KLF4 plays a promoter role in tumorigenesis and development, or tumor suppressor as a context-dependent anti- and pro-inflammatory factor. KLF4 over-expression increases CD8+T cell differentiation and enhances the antitumor immunity. This review aims to provide information about the relationship of KLF4 in immunity with tumors and to guide the future study.
Collapse
Affiliation(s)
- Yunjie Ju
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bryan James Mathis
- Clinical Research Manuscript Elevation Service, University of Tsukuba Institute of Medicine, Tsukuba, Japan
| | - Ying Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Elmitwalli O, Darwish R, Al-Jabery L, Algahiny A, Roy S, Butler AE, Hasan AS. The Emerging Role of p21 in Diabetes and Related Metabolic Disorders. Int J Mol Sci 2024; 25:13209. [PMID: 39684919 DOI: 10.3390/ijms252313209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In the context of cell cycle inhibition, anti-proliferation, and the dysregulation observed in certain cancer pathologies, the protein p21 assumes a pivotal role. p21 links DNA damage responses to cellular processes such as apoptosis, senescence, and cell cycle arrest, primarily functioning as a regulator of the cell cycle. However, accumulating empirical evidence suggests that p21 is both directly and indirectly linked to a number of different metabolic processes. Intriguingly, recent investigations indicate that p21 significantly contributes to the pathogenesis of diabetes. In this review, we present a comprehensive evaluation of the scientific literature regarding the involvement of p21 in metabolic processes, diabetes etiology, pancreatic function, glucose homeostasis, and insulin resistance. Furthermore, we provide an encapsulated overview of therapies that target p21 to alleviate metabolic disorders. A deeper understanding of the complex interrelationship between p21 and diabetes holds promise for informing current and future therapeutic strategies to address this rapidly escalating health crisis.
Collapse
Affiliation(s)
- Omar Elmitwalli
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Radwan Darwish
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Lana Al-Jabery
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ahmed Algahiny
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Sornali Roy
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Alexandra E Butler
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ammar S Hasan
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| |
Collapse
|
6
|
Kasirzadeh S, Lenjisa JL, Wang S. Targeting CDK2 to combat drug resistance in cancer therapy. Future Oncol 2024; 20:3325-3341. [PMID: 39469865 PMCID: PMC11633421 DOI: 10.1080/14796694.2024.2416382] [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: 08/14/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Drug resistance remains a major obstacle in cancer treatment, leading to treatment failures and high mortality rates. Despite advancements in therapies, overcoming resistance requires a deeper understanding of its mechanisms. This review highlights CDK2's pivotal role in both intrinsic and acquired resistance, and its potential as a therapeutic target. Cyclin E upregulation, which partners with CDK2, is linked to poor prognosis and resistance across various cancers. Specifically, amplifications of CCNE1/CCNE2 are associated with resistance to targeted therapies, immunotherapy, endocrine therapies and chemo/radiotherapy. Given CDK2's involvement in resistance mechanisms, investigating its role presents promising opportunities for developing novel strategies to combat resistance and improve treatment outcomes.
Collapse
Affiliation(s)
- Sara Kasirzadeh
- Drug Discovery & Development, Clinical & Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Jimma Likisa Lenjisa
- Drug Discovery & Development, Clinical & Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Shudong Wang
- Drug Discovery & Development, Clinical & Health Sciences, University of South Australia, Adelaide, 5000, Australia
| |
Collapse
|
7
|
Huang SY, Yu TS, Lin JH, Liu WH, Chung CA, Cheng YC. Stable laminar shear stress induces G1 cell cycle arrest and autophagy in urothelial carcinoma by a torque sensor-coupled cone-and-plate device. Eur J Cell Biol 2024; 103:151451. [PMID: 39217678 DOI: 10.1016/j.ejcb.2024.151451] [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: 01/18/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
The microenvironments of urinary systems play crucial roles in the development and metastasis of cancers due to their generation of complex temporal and spatial fluidic profiles. Because of their versatility in creating desired biomimetic flow, cone-and-plate bioreactors offer great potential for bladder cancer research. In this study, we construct a biocompatible cone-and-plate device coupled with a torque sensor, enabling the application and real-time monitoring of stable shear stress up to 50 dyne/cm². Under a stable shear stress stimulation at 12 dyne/cm2, bladder cancer cell BFTC-905 is arrested at the G1 phase with decreased cell proliferation after 24-hour treatment. This effect is associated with increased cyclin-dependent kinase inhibitors p21 and p27, inhibiting cyclin D1/CDK4 complex with dephosphorylation of serine 608 on the retinoblastoma protein. Consequently, an increase in cyclin D3 and decreases in cyclin A2 and cyclin E2 are observed. Moreover, we demonstrate that the shear stress stimulation upregulates the expression of autophagy-related proteins Beclin-1, LC3B-I and LC3B-II, while caspase cleavages are not activated under the same condition. The design of this system and its application shed new light on flow-induced phenomena in the study of urothelial carcinomas.
Collapse
Affiliation(s)
- Sheng-Yuan Huang
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, New Taipei City, Taiwan
| | - Tien-Ssu Yu
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan
| | - Jiun-Han Lin
- Department of Industrial Technology, Ministry of Economic Affairs, Taipei, Taiwan; Food Industry Research and Development Institute, Hsinchu City, Taiwan
| | - Wei-Hung Liu
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan
| | - Chih-Ang Chung
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan.
| | - Yu-Che Cheng
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, New Taipei City, Taiwan; Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
8
|
He K, Zhou D, Pu Z, Chen S, Shen Y, Zhao S, Qian X, Hu Q, Wu X, Xie Z, Xu X. Cellular Senescence in Acute Liver Injury: What Happens to the Young Liver? Aging Dis 2024; 16:1347-1362. [PMID: 38913043 PMCID: PMC12096906 DOI: 10.14336/ad.2024.0586] [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: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Cellular senescence, characterized by irreversible cell cycle arrest, not only exists in age-related physiological states, but has been found to exist in various diseases. It plays a crucial role in both physiological and pathological processes and has become a trending topic in global research in recent years. Acute liver injury (ALI) has a high incidence worldwide, and recent studies have shown that hepatic senescence can be induced following ALI. Therefore, we reviewed the significance of cellular senescence in ALI. To minimize the potential confounding effects of aging on cellular senescence and ALI outcomes, we selected studies involving young individuals to identify the characteristics of senescent cells, the value of cellular senescence in liver repair, its regulation mechanisms in ALI, its potential as a biomarker for ALI, the prospect of treatment, and future research directions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Datta I, Bangi E. Senescent cells and macrophages cooperate through a multi-kinase signaling network to promote intestinal transformation in Drosophila. Dev Cell 2024; 59:566-578.e3. [PMID: 38309266 PMCID: PMC10939848 DOI: 10.1016/j.devcel.2024.01.009] [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: 06/06/2023] [Revised: 11/21/2023] [Accepted: 01/12/2024] [Indexed: 02/05/2024]
Abstract
Cellular senescence is a conserved biological process that plays a crucial and context-dependent role in cancer. The highly heterogeneous and dynamic nature of senescent cells and their small numbers in tissues make in vivo mechanistic studies of senescence challenging. As a result, how multiple senescence-inducing signals are integrated in vivo to drive senescence in only a small number of cells is unclear. Here, we identify cells that exhibit multiple features of senescence in a Drosophila model of intestinal transformation, which emerge in response to concurrent activation of AKT, JNK, and DNA damage signaling within transformed tissue. Eliminating senescent cells, genetically or by treatment with senolytic compounds, reduces overgrowth and improves survival. We find that senescent cells promote tumorigenesis by recruiting Drosophila macrophages to the transformed tissue, which results in non-autonomous activation of JNK signaling. These findings identify senescent cell-macrophage interactions as an important driver of epithelial transformation.
Collapse
Affiliation(s)
- Ishwaree Datta
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA
| | - Erdem Bangi
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA.
| |
Collapse
|
10
|
Ooi LC, Ho V, Zhu JZ, Lim S, Chung L, Abubakar A, Rutland T, Chua W, Ng W, Lee M, Morgan M, MacKenzie S, Lee CS. p21 as a Predictor and Prognostic Indicator of Clinical Outcome in Rectal Cancer Patients. Int J Mol Sci 2024; 25:725. [PMID: 38255799 PMCID: PMC10815780 DOI: 10.3390/ijms25020725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/25/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
The cell cycle plays a key and complex role in the development of human cancers. p21 is a potent cyclin-dependent kinase inhibitor (CDKI) involved in the promotion of cell cycle arrest and the regulation of cellular senescence. Altered p21 expression in rectal cancer cells may affect tumor cells' behavior and resistance to neoadjuvant and adjuvant therapy. Our study aimed to ascertain the relationship between the differential expression of p21 in rectal cancer and patient survival outcomes. Using tissue microarrays, 266 rectal cancer specimens were immunohistochemically stained for p21. The expression patterns were scored separately in cancer cells retrieved from the center and the periphery of the tumor; compared with clinicopathological data, tumor regression grade (TRG), disease-free, and overall survival. Negative p21 expression in tumor periphery cells was significantly associated with longer overall survival upon the univariate (p = 0.001) and multivariable analysis (p = 0.003, HR = 2.068). Negative p21 expression in tumor periphery cells was also associated with longer disease-free survival in the multivariable analysis (p = 0.040, HR = 1.769). Longer overall survival times also correlated with lower tumor grades (p= 0.011), the absence of vascular and perineural invasion (p = 0.001; p < 0.005), the absence of metastases (p < 0.005), and adjuvant treatment (p = 0.009). p21 expression is a potential predictive and prognostic biomarker for clinical outcomes in rectal cancer patients. Negative p21 expression in tumor periphery cells demonstrated significant association with longer overall survival and disease-free survival. Larger prospective studies are warranted to investigate the ability of p21 to identify rectal cancer patients who will benefit from neoadjuvant and adjuvant therapy.
Collapse
Affiliation(s)
- Li Ching Ooi
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (L.C.O.); (J.Z.Z.); (T.R.); (C.S.L.)
| | - Vincent Ho
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Jing Zhou Zhu
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (L.C.O.); (J.Z.Z.); (T.R.); (C.S.L.)
| | - Stephanie Lim
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, NSW 2560, Australia
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Liping Chung
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Askar Abubakar
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
| | - Tristan Rutland
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (L.C.O.); (J.Z.Z.); (T.R.); (C.S.L.)
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Discipline of Medical Oncology, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Weng Ng
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Matthew Morgan
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Scott MacKenzie
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Department of Colorectal Surgery, Liverpool Hospital, Liverpool, NSW 2170, Australia;
| | - Cheok Soon Lee
- Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW 2170, Australia; (L.C.O.); (J.Z.Z.); (T.R.); (C.S.L.)
- School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia; (L.C.); (A.A.); (W.C.); (S.M.)
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia;
- Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| |
Collapse
|
11
|
Wu J, Huang X, Li X, Zhou H, Chen X, Chen Y, Guo Y, Huang J, Huang H, Huang Z, Chen G, Yang Z, Zhang J, Su W. Suppression of the long non-coding RNA LINC01279 triggers autophagy and apoptosis in lung cancer by regulating FAK and SIN3A. Discov Oncol 2024; 15:3. [PMID: 38168833 PMCID: PMC10761653 DOI: 10.1007/s12672-023-00855-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Long non-coding RNAs play critical roles in the development of lung cancer by functioning as tumor suppressors or oncogenes. Changes in the expression of LINC01279 have been associated with cell differentiation and human diseases. However, the mechanism underlying LINC01279 activity in tumorigenesis is not clear. Here, we analyzed the function of LINC01279 in lung adenocarcinoma using clinical samples, xenografts, and non-small-cell lung cancer cell lines. We found that LINC01279 is highly expressed in lung adenocarcinoma and may be considered as a predictive factor for this cancer. Knockdown of LINC01279 prevents tumor growth in xenografts and in cancer cell lines by activating autophagy and apoptosis. Molecularly, we revealed that LINC01279 regulates the expression of focal adhesion kinase and extracellular-regulated kinase signaling. In addition, it complexes with and stabilizes the transcriptional co-repressor SIN3A protein. Suppression of focal adhesion kinase and SIN3A also induces apoptosis and prevents tumor progression, suggesting that they may at least in part mediate the oncogenic activity of LINC01279. These results identify LINC01279 as a possible oncogene that plays an important role in the development of lung cancer. Our findings provide insights into the mechanism underlying LINC01279-mediated oncogenesis of lung adenocarcinoma. They may help to discover potential therapeutic targets for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Jiancong Wu
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaobi Huang
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaofang Li
- Center for Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Honglian Zhou
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaorao Chen
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongyang Chen
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yudong Guo
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jian Huang
- Department of Thoracic Surgery, Maoming People's Hospital, Maoming, China
| | - Hanqing Huang
- Department of Thoracic Surgery, Maoming People's Hospital, Maoming, China
| | - Zhong Huang
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guoan Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Zhixiong Yang
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Jian Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Wenmei Su
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
12
|
Zhang Z, Wang Z, Fan H, Li J, Ding J, Zhou G, Yuan C. The Indispensable Roles of GMDS and GMDS-AS1 in the Advancement of Cancer: Fucosylation, Signal Pathway and Molecular Pathogenesis. Mini Rev Med Chem 2024; 24:1712-1722. [PMID: 38591197 DOI: 10.2174/0113895575285276240324080234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Fucosylation is facilitated by converting GDP-mannose to GDP-4-keto-6-deoxymannose, which GDP-mannose 4,6-dehydratase, a crucial enzyme in the route, carries out. One of the most prevalent glycosylation alterations linked to cancer has reportedly been identified as fucosylation. There is mounting evidence that GMDS is intimately linked to the onset and spread of cancer. Furthermore, the significance of long-chain non-coding RNAs in the development and metastasis of cancer is becoming more well-recognized, and the regulatory mechanism of lncRNAs has emerged as a prominent area of study in the biological sciences. GMDS-AS1, an antisense RNA of GMDS, was discovered to have the potential to be an oncogene. We have acquired and analyzed relevant data to understand better how GMDS-AS1 and its lncRNA work physiologically and in tumorigenesis and progression. Additionally, we have looked into the possible effects of these molecules on cancer treatment approaches and patient outcomes. The physiological roles and putative processes of GMDS and lncRNA GMDS-AS1 throughout the development and progression of tumors have been assembled and examined. We also examined how these chemicals might affect patient prognosis and cancer therapy approaches. GMDS and GMDS-AS1 were determined to be research subjects by searching and gathering pertinent studies using the PubMed system. The analysis of these research articles demonstrated the close relationship between GMDS and GMDS-AS1 and tumorigenesis and the factors that influence them. GMDS plays a vital role in regulating fucosylation. The related antisense gene GMDS-AS1 affects the biological behaviors of cancer cells through multiple pathways, including the key processes of proliferation, migration, invasion, and apoptosis, providing potential biomarkers and therapeutic targets for cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Ziyan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Zhuowei Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Hong Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiayi Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiaqi Ding
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China
- Yichang Hospital of Traditional Chinese Medicine, Yichang 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| |
Collapse
|
13
|
Patricelli C, Lehmann P, Oxford JT, Pu X. Doxorubicin-induced modulation of TGF-β signaling cascade in mouse fibroblasts: insights into cardiotoxicity mechanisms. Sci Rep 2023; 13:18944. [PMID: 37919370 PMCID: PMC10622533 DOI: 10.1038/s41598-023-46216-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity has been widely observed, yet the specific impact on cardiac fibroblasts is not fully understood. Additionally, the modulation of the transforming growth factor beta (TGF-β) signaling pathway by DOX remains to be fully elucidated. This study investigated DOX's ability to modulate the expression of genes and proteins involved in the TGF-β signaling cascade in mouse fibroblasts from two sources by assessing the impact of DOX treatment on TGF-β inducible expression of pivotal genes and proteins within fibroblasts. Mouse embryonic fibroblasts (NIH3T3) and mouse primary cardiac fibroblasts (CFs) were treated with DOX in the presence of TGF-β1 to assess changes in protein levels by western blot and changes in mRNA levels by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Our results revealed a dose-dependent reduction in cellular communication network factor 2 (CCN2) protein levels upon DOX treatment in both NIH3T3 and CFs, suggesting an antifibrotic activity by DOX in these fibroblasts. However, DOX only inhibited the TGF-β1 induced expression of COL1 in NIH3T3 cells but not in CFs. In addition, we observed that DOX treatment reduced the expression of BMP1 in NIH3T3 but not primary cardiac fibroblasts. No significant changes in SMAD2 protein expression and phosphorylation in either cells were observed after DOX treatment. Finally, DOX inhibited the expression of Atf4 gene and increased the expression of Cdkn1a, Id1, Id2, Runx1, Tgfb1, Inhba, Thbs1, Bmp1, and Stat1 genes in NIH3T3 cells but not CFs, indicating the potential for cell-specific responses to DOX and its modulation of the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Conner Patricelli
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID, 83725-1512, USA
| | - Parker Lehmann
- Idaho College of Osteopathic Medicine, Meridian, ID, 83642-8046, USA
| | - Julia Thom Oxford
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID, 83725-1512, USA
- Biomolecular Research Center, Boise State University, Boise, ID, 83725-1511, USA
- Department of Biological Sciences, Boise State University, Boise, ID, 83725-1515, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID, 83725-1511, USA.
- Department of Biological Sciences, Boise State University, Boise, ID, 83725-1515, USA.
| |
Collapse
|
14
|
Cheng WY, Shen CC, Liang YJ, Chiao MT, Yang YC, Hsieh WY, Lin CH, Chen JP. Polymorphism at codon 31 of CDKN1A (p21) as a predictive factor for bevacizumab therapy in glioblastoma multiforme. BMC Cancer 2023; 23:886. [PMID: 37730565 PMCID: PMC10510274 DOI: 10.1186/s12885-023-11400-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023] Open
Abstract
Glioblastoma (GBM), a prevalent and malignant brain tumor, poses a challenge in surgical resection due to its invasive nature within the brain parenchyma. CDKN1A (p21, Waf-1), a cyclin-dependent kinase inhibitor, plays a pivotal role in regulating cell growth arrest, terminal differentiation, and apoptosis. The existence of natural variants of CDKN1A has been associated with specific cancer types. In this retrospective study, our objective was to identify polymorphic variants of CDKN1A, specifically c.93C > A (codon 31 Ser31Arg), and investigate its potential impact within the scope of bevacizumab therapy for glioblastoma multiforme. This study involved a cohort of 139 unrelated adult Chinese GBM patients in Taiwan. Genomic DNA extracted from tumor samples was utilized for genotyping using the polymerase chain reaction (PCR) restriction fragment length polymorphism method (PCR-RFLP analysis). Through unconditional logistic regression analysis, odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were calculated. Our findings unveiled that among these GBM patients, the distribution of codon 31 polymorphisms was as follows: 23.02% were Serine homozygotes (Ser/Ser), 27.34% were Arginine homozygotes (Arg/Arg), and 49.64% were Serine/Arginine heterozygotes (Ser/Arg). While CDKN1A c.93C > A polymorphisms did not exhibit a direct association with overall survival in GBM patients, noteworthy survival benefits emerged among individuals with Arg/Arg and Arg/Ser genotypes who received combined concurrent chemoradiotherapy (CCRT) and bevacizumab treatment compared to those who underwent CCRT alone. Our findings indicate a significant involvement of the CDKN1A c.93C > A polymorphism in the development and onset of GBM, offering potential implications for the early prognostication of bevacizumab therapy outcomes.
Collapse
Affiliation(s)
- Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan.
- Department of Physical Therapy, Hung Kuang University, Taichung city, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung city, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung city, Taiwan.
| | - Chiung-Chyi Shen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
- Department of Physical Therapy, Hung Kuang University, Taichung city, Taiwan
- Basic Medical Education, Central Taiwan University of Science and Technology, Taichung city, Taiwan
| | - Yea-Jiuen Liang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
| | - Ming-Tsang Chiao
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
| | - Yi-Chin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
| | - Wan-Yu Hsieh
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
| | - Cheng-Hui Lin
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung city, Taiwan
| | - Jun-Peng Chen
- Biostatistics Task Force, Taichung Veterans General Hospital, Taichung city, Taiwan
| |
Collapse
|
15
|
Wang Q, Zhao M, Zhang T, Zhang B, Zheng Z, Lin Z, Zhou S, Zheng D, Chen Z, Zheng S, Zhang Y, Lin X, Dong R, Chen J, Qian H, Hu X, Zhuang Y, Zhang Q, Jiang S, Ma Y. Comprehensive analysis of ferroptosis-related genes in immune infiltration and prognosis in multiple myeloma. Front Pharmacol 2023; 14:1203125. [PMID: 37608887 PMCID: PMC10440437 DOI: 10.3389/fphar.2023.1203125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Abstract
Background: One particular type of cellular death that is known as ferroptosis is caused by the excessive lipid peroxidation. It is a regulated form of cell death that can affect the response of the tumor cells. Currently, it is not known if the presence of this condition can affect the prognosis of patients with multiple myeloma (MM). Methods: In this study, we studied the expression differences and prognostic value of ferroptosis-related genes (FRGs) in MM, and established a ferroptosis risk scoring model. In order to improve the prediction accuracy and clinical applicability, a nomogram was also established. Through gene enrichment analysis, pathways closely related to high-risk groups were identified. We then explored the differences in risk stratification in drug sensitivity and immune patterns, and evaluated their value in prognostic prediction and treatment response. Lastly, we gathered MM cell lines and samples from patients to confirm the expression of marker FRGs using quantitative real-time PCR (qRT-PCR). Results: The ability to predict the survival of MM patients is a challenging issue. Through the use of a risk model derived from ferroptosis, we were able to develop a more accurate prediction of the disease's prognosis. They were then validated by a statistical analysis, which showed that the model is an independent factor in the prognosis of MM. Patients of high ferroptosis risk scores had a much worse chance of survival than those in the low-risk groups. The calibration and power of the nomogram were also strong. We noted that the link between the ferroptosis risk score and the clinical treatment was suggested by the FRG's significant correlation with the immune checkpoint genes and the medication sensitivity. We validated the predictive model using qRT-PCR. Conclusion: We demonstrated the association between FRGs and MM, and developed a new risk model for prognosis in MM patients. Our study sheds light on the potential clinical relevance of ferroptosis in MM and highlights its potential as a therapeutic target for patients with this disease.
Collapse
Affiliation(s)
- Quanqiang Wang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Misheng Zhao
- Department of Clinical Laboratory, Wenzhou People’s Hospital, Wenzhou, China
| | - Tianyu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bingxin Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziwei Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhili Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shujuan Zhou
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dong Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zixing Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanru Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rujiao Dong
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Honglan Qian
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Hu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianying Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyong Ma
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, Zhejiang, China
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Patricelli C, Lehmann P, Oxford JT, Pu X. Doxorubicin-Induced Modulation of TGF-β Signaling Cascade in Mouse Fibroblasts: Insights into Cardiotoxicity Mechanisms. RESEARCH SQUARE 2023:rs.3.rs-3186393. [PMID: 37546862 PMCID: PMC10402200 DOI: 10.21203/rs.3.rs-3186393/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Doxorubicin (DOX)-induced cardiotoxicity has been widely observed, yet the specific impact on cardiac fibroblasts is not fully understood. Additionally, the modulation of the transforming growth factor beta (TGF-β) signaling pathway by DOX remains to be fully elucidated. This study investigated DOX's ability to modulate the expression of genes and proteins involved in the TGF-β signaling cascade in mouse fibroblasts from two sources by assessing the impact of DOX treatment on TGF-β inducible expression of pivotal genes and proteins within fibroblasts. Mouse embryonic fibroblasts (NIH3T3) and mouse primary cardiac fibroblasts (CFs) were treated with DOX in the presence of TGF-β1 to assess changes in protein levels by western blot and changes in mRNA levels by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Our results revealed a dose-dependent reduction in cellular communication network factor 2 (CCN2) protein levels upon DOX treatment in both NIH3T3 and CFs. Moreover, we observed that DOX inhibited the TGF-β1 induced expression of BMP1 in NIH3T3 cells, while BMP1 levels remained high in CFs, and that TGF-β1 induces the phosphorylation of SMAD2 in both NIH3T3 cells and CFs. While DOX treatment diminished the extent of phosphorylation, the reduction did not reach statistical significance. DOX also inhibited the TGF-β1 induced expression of COL1 in NIH3T3 cells and CFs. Finally, DOX inhibited the TGF-β1 induced expression of Atf4 and increased the expression of Cdkn1a, Id1, Id2, Runx1, Tgfb1, Inhba, Thbs1, Bmp1, and Stat1 in NIH3T3 cells but not CFs, indicating the potential for cell-specific responses to DOX and its modulation of the TGF-β signaling pathway. Understanding the underlying mechanisms of the ability of DOX to modulate gene expression and signaling pathways in fibroblasts holds promise for future development of targeted therapeutic strategies to mitigate DOX-induced cardiotoxicity specifically affecting CFs.
Collapse
|
17
|
Datta I, Bangi E. Senescent cells and macrophages cooperate through a multi-kinase signaling network to promote intestinal transformation in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540869. [PMID: 37292988 PMCID: PMC10245684 DOI: 10.1101/2023.05.15.540869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cellular senescence is a conserved biological process essential for embryonic development, tissue remodeling, repair, and a key regulator of aging. Senescence also plays a crucial role in cancer, though this role can be tumor-suppressive or tumor-promoting, depending on the genetic context and the microenvironment. The highly heterogeneous, dynamic, and context-dependent nature of senescence-associated features and the relatively small numbers of senescent cells in tissues makes in vivo mechanistic studies of senescence challenging. As a result, which senescence-associated features are observed in which disease contexts and how they contribute to disease phenotypes remain largely unknown. Similarly, the specific mechanisms by which various senescence-inducing signals are integrated in vivo to induce senescence and why some cells become senescent while their immediate neighbors do not are unclear. Here, we identify a small number of cells that exhibit multiple features of senescence in a genetically complex model of intestinal transformation we recently established in the developing Drosophila larval hindgut epithelium. We demonstrate that these cells emerge in response to concurrent activation of AKT, JNK, and DNA damage response pathways within transformed tissue. Eliminating senescent cells, genetically or by treatment with senolytic compounds, reduces overgrowth and improves survival. We find that this tumor-promoting role is mediated by Drosophila macrophages recruited to the transformed tissue by senescent cells, which results in non-autonomous activation of JNK signaling within the transformed epithelium. These findings emphasize complex cell-cell interactions underlying epithelial transformation and identify senescent cell-macrophage interactions as a potential druggable node in cancer.
Collapse
Affiliation(s)
- Ishwaree Datta
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA
| | - Erdem Bangi
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA
| |
Collapse
|
18
|
Na Y, Hall A, Yu Y, Hu L, Choi K, Burgard JA, Szabo S, Huang G, Ratner N, Wu J. Runx1/3-driven adaptive endoplasmic reticulum stress pathways contribute to neurofibromagenesis. Oncogene 2023; 42:1038-1047. [PMID: 36759572 PMCID: PMC10194627 DOI: 10.1038/s41388-023-02620-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023]
Abstract
Neurofibromatosis type 1 (NF1) patients are predisposed to develop plexiform neurofibromas (PNFs). Three endoplasmic reticulum (ER) stress response pathways restore cellular homeostasis. The unfolded protein response (UPR) sensors contribute to tumor initiation in many cancers. We found that all three UPR pathways were activated in mouse and human PNFs, with protein kinase RNA [PKR]-like ER kinase (PERK) the most highly expressed. We tested if neurofibroma cells adapt to ER stress, leading to their growth. Pharmacological or genetic inhibition of PERK reduced mouse neurofibroma-sphere number, and genetic inhibition in PERK in Schwann cell precursors (SCPs) decreased tumor-like lesion numbers in a cell transplantation model. Further, in a PNF mouse model, deletion of PERK in Schwann cells (SCs) and SCPs reduced tumor size, number, and increased survival. Mechanistically, loss of Nf1 activated PERK-eIF2α-ATF4 signaling and increased ATF4 downstream target gene p21 translocation from nucleus to cytoplasm. This nucleus-cytoplasm translocation was mediated by exportin-1. Runx transcriptionally activated ribosome gene expression and increased protein synthesis to allow SCs to adapt to ER stress and tumor formation. We propose that targeting proteostasis might provide cytotoxic and/or potentially durable novel therapy for PNFs.
Collapse
Affiliation(s)
- Youjin Na
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Ashley Hall
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Yanan Yu
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- College of Life Science, Xuzhou Medical University, 221004, Jiangsu, P. R. China
| | - Liang Hu
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Jake A Burgard
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Sara Szabo
- Department of Pediatrics and Department of Pediatric Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Gang Huang
- Department of Cell Systems & Anatomy and Department of Pathology & Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
- Department of Pathology & Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, Mays Cancer Center at UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
19
|
Maheshwari M, Yadav N, Hasanain M, Pandey P, Sahai R, Choyal K, Singh A, Nengroo MA, Saini KK, Kumar D, Mitra K, Datta D, Sarkar J. Inhibition of p21 activates Akt kinase to trigger ROS-induced autophagy and impacts on tumor growth rate. Cell Death Dis 2022; 13:1045. [PMID: 36522339 PMCID: PMC9755229 DOI: 10.1038/s41419-022-05486-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Owing to its ability to induce cellular senescence, inhibit PCNA, and arrest cell division cycle by negatively regulating CDKs as well as being a primary target of p53, p21 is traditionally considered a tumor suppressor. Nonetheless, several reports in recent years demonstrated its pro-oncogenic activities such as apoptosis inhibition by cytosolic p21, stimulation of cell motility, and promoting assembly of cyclin D-CDK4/6 complex. These opposing effects of p21 on cell proliferation, supported by the observations of its inconsistent expression in human cancers, led to the emergence of the concept of "antagonistic duality" of p21 in cancer progression. Here we demonstrate that p21 negatively regulates basal autophagy at physiological concentration. Akt activation, upon p21 attenuation, driven ROS accumulation appears to be the major underlying mechanism in p21-mediated modulation of autophagy. We also find p21, as a physiological inhibitor of autophagy, to have oncogenic activity during early events of tumor development while its inhibition favors survival and growth of cancer cells in the established tumor. Our data, thereby, reveal the potential role of autophagy in antagonistic functional duality of p21 in cancer.
Collapse
Affiliation(s)
- Mayank Maheshwari
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Nisha Yadav
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Mohammad Hasanain
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Praveen Pandey
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Rohit Sahai
- grid.418363.b0000 0004 0506 6543Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Kuldeep Choyal
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Akhilesh Singh
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Mushtaq A. Nengroo
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Krishan K. Saini
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Deepak Kumar
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Kalyan Mitra
- grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India ,grid.418363.b0000 0004 0506 6543Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Dipak Datta
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Jayanta Sarkar
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| |
Collapse
|
20
|
Mir KB, Faheem MM, Ahmad SM, Rasool JU, Amin T, Chakraborty S, Bhagat M, Ahmed Z, Ali A, Goswami A. β-(4-fluorobenzyl) Arteannuin B induced interaction of ATF-4 and C/EBPβ mediates the transition of breast cancer cells from autophagy to senescence. Front Oncol 2022; 12:1013500. [PMID: 36465376 PMCID: PMC9713483 DOI: 10.3389/fonc.2022.1013500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/17/2022] [Indexed: 08/06/2023] Open
Abstract
ATF-4 is a master regulator of transcription of genes essential for cellular-adaptive function. In response to the quantum and duration of stress, ATF-4 diligently responds to both pro-apoptotic and pro-survival signals converging into either autophagy or apoptosis/senescence. Despite emerging cues implying a relationship between autophagy and senescence, how these two processes are controlled remains unknown. Herein, we demonstrate β-(4-fluorobenzyl) Arteannuin B (here after Arteannuin 09), a novel semisynthetic derivative of Arteannuin B, as a potent ER stress inducer leading to the consistent activation of ATF-4. Persistent ATF-4 expression at early time-points facilitates the autophagy program and consequently by upregulating p21 at later time-points, the signaling is shifted towards G2/M cell cycle arrest. As bZIP transcription factors including ATF-4 are obligate dimers, and because ATF-4 homodimers are not highly stable, we hypothesized that ATF-4 may induce p21 expression by physically interacting with another bZIP family member i.e., C/EBPβ. Our co-immunoprecipitation and co-localization studies demonstrated that ATF-4 is principally responsible for the autophagic potential of Arteannuin 09, while as, induction of both ATF-4 and C/EBPβ is indispensable for the p21 regulated-cell cycle arrest. Interestingly, inhibition of autophagy signaling switches the fate of Arteannuin 09 treated cells from senescence to apoptosis. Lastly, our data accomplished that Arteannuin 09 is a potent inhibitor of tumor growth and inducer of premature senescence in vivo.
Collapse
Affiliation(s)
- Khalid Bashir Mir
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
| | - Mir Mohd Faheem
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
- School of Biotechnology, University of Jammu, Jammu, India
| | - Syed Mudabir Ahmad
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
| | - Javeed Ur Rasool
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Natural Product and Medicinal Chemistry Division, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu, India
| | - Tanzeeba Amin
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
| | | | | | - Zabeer Ahmed
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
| | - Asif Ali
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Natural Product and Medicinal Chemistry Division, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Integrative Medicine, Jammu, India
- Division of Medicinal and Process Chemistry, Council of Scientific & Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Anindya Goswami
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
- Pharmacology Division, Council of Scientific & Industrial Research (CSIR)-Indian Indian Institute of Integrative Medicine, Jammu, India
| |
Collapse
|
21
|
Nanić L, Cedilak A, Vidaček NŠ, Gruber F, Huzak M, Bader M, Rubelj I. In Vivo Skin Regeneration and Wound Healing Using Cell Micro-Transplantation. Pharmaceutics 2022; 14:pharmaceutics14091955. [PMID: 36145701 PMCID: PMC9501230 DOI: 10.3390/pharmaceutics14091955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Background: The accumulation of senescent cells in tissues alters tissue homeostasis and affects wound healing. It is also considered to be the main contributing factor to aging. In addition to losing their ability to divide, senescent cells exert detrimental effects on surrounding tissues through their senescence-associated secretory phenotype (SASP). They also affect stem cells and their niche, reducing their capacity to divide which increasingly reduces tissue regenerative capacity over time. The aim of our study was to restore aged skin by increasing the fraction of young cells in vivo using a young cell micro-transplantation technique on Fischer 344 rats. Employing the same technique, we also used wild-type skin fibroblasts and stem cells in order to heal Dominant Dystrophic Epidermolysis Bulosa (DDEB) wounds and skin blistering. Results: We demonstrate that implantation of young fibroblasts restores cell density, revitalizes cell proliferation in the dermis and epidermis, rejuvenates collagen I and III matrices, and boosts epidermal stem cell proliferation in rats with advancing age. We were also able to reduce blistering in DDEB rats by transplantation of skin stem cells but not skin fibroblasts. Conclusions: Our intervention proves that a local increase of young cells in the dermis changes tissue homeostasis well enough to revitalize the stem cell niche, ensuring overall skin restoration and rejuvenation as well as healing DDEB skin. Our method has great potential for clinical applications in skin aging, as well as for the treatment of various skin diseases.
Collapse
Affiliation(s)
- Lucia Nanić
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Andrea Cedilak
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Nikolina Škrobot Vidaček
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruder Boskovic Institute, 10000 Zagreb, Croatia
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Miljenko Huzak
- Department of Mathematics, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Institute for Biology, University of Lübeck, 23562 Lübeck, Germany
| | - Ivica Rubelj
- Laboratory for Molecular and Cellular Biology, Division of Molecular Biology, Ruder Boskovic Institute, 10000 Zagreb, Croatia
- Correspondence:
| |
Collapse
|
22
|
Jayasinghe R, Jayarajah U, Seneviratne S. Circulating Biomarkers in Predicting Pathological Response to Neoadjuvant Therapy for Colorectal Cancer. Biomark Med 2022. [DOI: 10.2174/9789815040463122010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating biomarkers show promise in the management of many cancers.
They have become the novel non-invasive approach to complement the current
strategies in colorectal cancer (CRC) management. Their ability in guiding diagnosis,
evaluating response to treatment, screening and prognosis is phenomenal, especially
when it comes to their minimally invasive nature. These “liquid biopsies,” which show
potential for replacing invasive surgical biopsies, provide useful information on the
primary and metastatic disease by providing an insight into cancer biology. Analysis of
blood and body fluids for circulating tumour DNA (ctDNA), carcinoembryonic antigen
(CEA), circulating tumour cells (CTC), or circulating micro RNA (miRNA) shows
potential for improving CRC management. Recognizing a predictive model to assess
response to neoadjuvant chemotherapy would help in better patient selection. This
review was conducted with the aim of outlining the use of circulatory biomarkers in
current practice and their effectiveness in the management of patients having CRC with
a focus on response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Ravindri Jayasinghe
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Umesh Jayarajah
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Sanjeewa Seneviratne
- Department of Surgery, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
23
|
Ungerleider K, Beck JA, Lissa D, Joruiz S, Horikawa I, Harris CC. Δ133p53α Protects Human Astrocytes from Amyloid-beta Induced Senescence and Neurotoxicity. Neuroscience 2022; 498:190-202. [PMID: 35716965 PMCID: PMC9420812 DOI: 10.1016/j.neuroscience.2022.06.004] [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: 09/03/2021] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Cellular senescence is an important contributor to aging and age-related diseases such as Alzheimer's disease (AD). Senescent cells are characterized by a durable cell proliferation arrest and the acquisition of a proinflammatory senescence-associated secretory phenotype (SASP), which participates in the progression of neurodegenerative disorders. Clearance of senescent glial cells in an AD mouse model prevented cognitive decline suggesting pharmacological agents targeting cellular senescence might provide novel therapeutic approaches for AD. Δ133p53α, a natural protein isoform of p53, was previously shown to be a negative regulator of cellular senescence in primary human astrocytes, with clinical implications from its diminished expression in brain tissues from AD patients. Here we show that treatment of proliferating human astrocytes in culture with amyloid-beta oligomers (Aβ), an endogenous pathogenic agent of AD, results in reduced expression of Δ133p53α, as well as induces the cells to become senescent and express proinflammatory SASP cytokines such as IL-6, IL-1β and TNFα. Our data suggest that Aβ-induced astrocyte cellular senescence is associated with accelerated DNA damage, and upregulation of full-length p53 and its senescence-inducing target gene p21WAF1. We also show that exogenously enhanced expression of Δ133p53α rescues human astrocytes from Aβ-induced cellular senescence and SASP through both protection from DNA damage and dominant-negative inhibition of full-length p53, leading to inhibition of Aβ-induced, astrocyte-mediated neurotoxicity. The results presented here demonstrate that Δ133p53α manipulation could modulate cellular senescence in the context of AD, possibly opening new therapeutic avenues.
Collapse
Affiliation(s)
- Kyra Ungerleider
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jessica A Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Comparative Pathobiology, Purdue University, West Layfette, IN 47907, USA
| | - Delphine Lissa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sebastien Joruiz
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Schirripa A, Sexl V, Kollmann K. Cyclin-dependent kinase inhibitors in malignant hematopoiesis. Front Oncol 2022; 12:916682. [PMID: 36033505 PMCID: PMC9403899 DOI: 10.3389/fonc.2022.916682] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The cell-cycle is a tightly orchestrated process where sequential steps guarantee cellular growth linked to a correct DNA replication. The entire cell division is controlled by cyclin-dependent kinases (CDKs). CDK activation is balanced by the activating cyclins and CDK inhibitors whose correct expression, accumulation and degradation schedule the time-flow through the cell cycle phases. Dysregulation of the cell cycle regulatory proteins causes the loss of a controlled cell division and is inevitably linked to neoplastic transformation. Due to their function as cell-cycle brakes, CDK inhibitors are considered as tumor suppressors. The CDK inhibitors p16INK4a and p15INK4b are among the most frequently altered genes in cancer, including hematopoietic malignancies. Aberrant cell cycle regulation in hematopoietic stem cells (HSCs) bears severe consequences on hematopoiesis and provokes hematological disorders with a broad array of symptoms. In this review, we focus on the importance and prevalence of deregulated CDK inhibitors in hematological malignancies.
Collapse
|
25
|
Sirtuin 4 Inhibits Prostate Cancer Progression and Metastasis by Modulating p21 Nuclear Translocation and Glutamate Dehydrogenase 1 ADP-Ribosylation. JOURNAL OF ONCOLOGY 2022; 2022:5498743. [PMID: 35847357 PMCID: PMC9283077 DOI: 10.1155/2022/5498743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/14/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022]
Abstract
Protein posttranslational modification regulates several biological mechanisms, including tumor progression. In this study, we show that the mitochondrial Sirtuin 4 (SIRT4), which has ADP-ribosylation activity, plays a role in prostate cancer (PCa) progression. Firstly, SIRT4 expression was verified in PCa tissues and cell lines by quantitative real-time PCR (qRT-PCR) and western blotting. Subsequently, we established stable PC-3 and 22rv1 cells that overexpressed SIRT4 and knocked down SIRT4, respectively. The functions of SIRT4 in PCa were explored through various phenotype experiments. The mechanism underlying the functions of SIRT4 was investigated through western blotting, immunoprecipitation, immunofluorescence, and nuclear and cytoplasmic extraction assays. We revealed that SIRT4 inhibited cell progression both in vivo and in vitro. Mechanistically, on the one hand, SIRT4 promoted the ADP-ribosylation of glutamate dehydrogenase 1 to inhibit the glutamine metabolism pathways. On the other hand, SIRT4 inhibited the phosphorylation of AKT, thereby affecting p21 phosphorylation and its cellular localization for cell cycle arrest. In conclusion, our study indicates that SIRT4 is directly associated with PCa progression and could be a novel target for PCa therapy.
Collapse
|
26
|
Debnath S, Sarkar A, Mukherjee DD, Ray S, Mahata B, Mahata T, Parida PK, Das T, Mukhopadhyay R, Ghosh Z, Biswas K. Eriodictyol mediated selective targeting of the TNFR1/FADD/TRADD axis in cancer cells induce apoptosis and inhibit tumor progression and metastasis. Transl Oncol 2022; 21:101433. [PMID: 35462210 PMCID: PMC9046888 DOI: 10.1016/j.tranon.2022.101433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/03/2022] Open
Abstract
While the anti-inflammatory activities of Eriodictyol, a plant-derived flavonoid is well-known, reports on its anti-cancer efficacy and selective cytotoxicity in cancer cells are still emerging. However, little is known regarding its mechanism of selective anti-cancer activities. Here, we show the mechanism of selective cytotoxicity of Eriodictyol towards cancer cells compared to normal cells. Investigation reveals that Eriodictyol significantly upregulates TNFR1 expression in tumor cells (HeLa and SK-RC-45) while sparing the normal cells (HEK, NKE and WI-38), which display negligible TNFR1 expression, irrespective of the absence or presence of Eriodictyol. Further investigation of the molecular events reveal that Eriodictyol induces apoptosis through expression of the pro-apoptotic DISC components leading to activation of the caspase cascade. In addition, CRISPR-Cas9 mediated knockout of TNFR1 completely blocks apoptosis in HeLa cells in response to Eriodictyol, confirming that Eriodictyol induced cancer cell apoptosis is indeed TNFR1-dependent. Finally, in vivo data demonstrates that Eriodictyol not only impedes tumor growth and progression, but also inhibits metastasis in mice implanted with 4T1 breast cancer cells. Thus, our study has identified Eriodictyol as a compound with high selectivity towards cancer cells through TNFR1 and suggests that it can be further explored for its prospect in cancer therapeutics.
Collapse
Affiliation(s)
- Shibjyoti Debnath
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Abhisek Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | | | - Subha Ray
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Barun Mahata
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Tarun Mahata
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Pravat K Parida
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Troyee Das
- The Bioinformatics Center, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Rupak Mukhopadhyay
- Department of Molecular Biology & Biotechnology, Tezpur University, Assam 784028
| | - Zhumur Ghosh
- The Bioinformatics Center, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Kaushik Biswas
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
27
|
Use of RNA-Seq and a Transgenic Mouse Model to Identify Genes Which May Contribute to Mutant p53-Driven Prostate Cancer Initiation. BIOLOGY 2022; 11:biology11020218. [PMID: 35205085 PMCID: PMC8869245 DOI: 10.3390/biology11020218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/03/2022]
Abstract
Simple Summary We use RNA-seq analysis to identify genes that may contribute to mutant p53-mediated prostate cancer initiation in a genetically engineered mouse model (B6.129S4-Trp53tm3.1Tyj/J). A total of 1378 differentially expressed genes, including wildtype p53 target genes (e.g. Cdkn1a, Bax, Bcl2, Kras, Mdm2), p53 gain-of-function-related genes (Mgmt, Id4), and prostate cancer-related genes (Cav-1, Raf1, Kras), were identified. Mice that were homozygous or heterozygous for the Trp53 R270H mutation developed grade one PIN lesions at 3 months and 5 months, respectively, whereas wildtype mice did not develop PIN. Immunohistochemical analysis revealed decreased levels of irradiation-mediated apoptosis in homozygous and heterozygous mice when compared to wildtype counterparts, and this aligned with observed differences in apoptosis-related gene expression. Abstract We previously demonstrated that the Trp53-R270H mutation can drive prostate cancer (CaP) initiation using the FVB.129S4 (Trp53tm3Tyj/wt); FVB.129S (Nkx3-1tm3(cre)Mmswt) genetically engineered mouse model (GEM). We now validate this finding in a different model (B6.129S4-Trp53tm3.1Tyj/J mice) and use RNA-sequencing (RNA-Seq) to identify genes which may contribute to Trp53 R270H-mediated prostate carcinogenesis. Wildtype (Trp53WT/WT), heterozygous (Trp53R270H/WT), and homozygous mice (Trp53R270H/R270H) were exposed to 5 Gy irradiation to activate and stabilize p53, and thereby enhance our ability to identify differences in transcriptional activity between the three groups of mice. Mouse prostates were harvested 6 h post-irradiation and processed for histological/immunohistochemistry (IHC) analysis or were snap-frozen for RNA extraction and transcriptome profiling. IHC analyses determined that presence of the Trp53-R270H mutation impacts apoptosis (lower caspase 3 activity) but not cell proliferation (Ki67). RNA-Seq analysis identified 1378 differentially expressed genes, including wildtype p53 target genes (E.g., Cdkn1a, Bax, Bcl2, Kras, Mdm2), p53 gain-of-function (GOF)-related genes (Mgmt, Id4), and CaP-related genes (Cav-1, Raf1, Kras). Further understanding the mechanisms which contribute to prostate carcinogenesis could allow for the development of improved preventive methods, diagnostics, and treatments for CaP.
Collapse
|
28
|
Liao Z, Yeo HL, Wong SW, Zhao Y. Cellular Senescence: Mechanisms and Therapeutic Potential. Biomedicines 2021; 9:1769. [PMID: 34944585 PMCID: PMC8698401 DOI: 10.3390/biomedicines9121769] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a complex and multistep biological process which cells can undergo in response to different stresses. Referring to a highly stable cell cycle arrest, cellular senescence can influence a multitude of biological processes-both physiologically and pathologically. While phenotypically diverse, characteristics of senescence include the expression of the senescence-associated secretory phenotype, cell cycle arrest factors, senescence-associated β-galactosidase, morphogenesis, and chromatin remodelling. Persistent senescence is associated with pathologies such as aging, while transient senescence is associated with beneficial programmes, such as limb patterning. With these implications, senescence-based translational studies, namely senotherapy and pro-senescence therapy, are well underway to find the cure to complicated diseases such as cancer and atherosclerosis. Being a subject of major interest only in the recent decades, much remains to be studied, such as regarding the identification of unique biomarkers of senescent cells. This review attempts to provide a comprehensive understanding of the diverse literature on senescence, and discuss the knowledge we have on senescence thus far.
Collapse
Affiliation(s)
- Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-17177 Stockholm, Sweden
| | - Han Lin Yeo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Siaw Wen Wong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore;
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
29
|
Virdis P, Marchesi I, Fiorentino FP, Migheli R, Sanna L, Bordoni V, Pintore G, Galleri G, Muroni MR, Bagella L, Fozza C, De Miglio MR, Podda L. Tomentosin a Sesquiterpene Lactone Induces Antiproliferative and Proapoptotic Effects in Human Burkitt Lymphoma by Deregulation of Anti- and Pro-Apoptotic Genes. Life (Basel) 2021; 11:life11111128. [PMID: 34833004 PMCID: PMC8623649 DOI: 10.3390/life11111128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
(1) Tomentosin is the most representative sesquiterpene lactone extracted by I. viscosa. Recently, it has gained particular attention in therapeutic oncologic fields due to its anti-tumor properties. (2) In this study, the potential anticancer features of tomentosin were evaluated on human Burkitt’s lymphoma (BL) cell line, treated with increasing tomentosin concentration for cytotoxicity screening. (3) Our data showed that both cell cycle arrest and cell apoptosis induction are responsible of the antiproliferative effects of tomentosin and may end in the inhibition of BL cell viability. Moreover, a microarray gene expression profile was performed to assess differentially expressed genes contributing to tomentosin activity. Seventy-five genes deregulated by tomentosin have been identified. Downregulated genes are enriched in immune-system pathways, and PI3K/AKT and JAK/STAT pathways which favor proliferation and growth processes. Importantly, different deregulated genes identified in tomentosin-treated BL cells are prevalent in molecular pathways known to lead to cellular death, specifically by apoptosis. Tomentosin-treatment in BL cells induces the downregulation of antiapoptotic genes such as BCL2A1 and CDKN1A and upregulation of the proapoptotic PMAIP1 gene. (4) Overall, our results suggest that tomentosin could be taken into consideration as a potential natural product with limited toxicity and relevant anti-tumoral activity in the therapeutic options available to BL patients.
Collapse
Affiliation(s)
- Patrizia Virdis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| | - Irene Marchesi
- Kitos Biotech Srls, Porto Conte Ricerche, 07100 Sassari, Italy; (I.M.); (F.P.F.)
| | | | - Rossana Migheli
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| | - Luca Sanna
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| | - Valentina Bordoni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.B.); (L.B.)
| | - Giorgio Pintore
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Grazia Galleri
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.B.); (L.B.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Claudio Fozza
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
- Correspondence: (C.F.); (M.R.D.M.)
| | - Maria Rosaria De Miglio
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
- Correspondence: (C.F.); (M.R.D.M.)
| | - Luigi Podda
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.V.); (R.M.); (L.S.); (G.G.); (M.R.M.); (L.P.)
| |
Collapse
|
30
|
Vivekanandhan S, Madamsetty VS, Angom RS, Dutta SK, Wang E, Caulfield T, Pletnev AA, Upstill-Goddard R, Asmann YW, Chang D, Spaller MR, Mukhopadhyay D. Role of PLEXIND1/TGFβ Signaling Axis in Pancreatic Ductal Adenocarcinoma Progression Correlates with the Mutational Status of KRAS. Cancers (Basel) 2021; 13:cancers13164048. [PMID: 34439202 PMCID: PMC8393884 DOI: 10.3390/cancers13164048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is among the most lethal cancers. The expression of PLEXIND1, a receptor, is upregulated in many cancers (including pancreatic cancer). Traditionally, PLEXIND1 is known to be involved in neuron development and mediate semaphorin signaling. However, its role and signaling in cancer is not fully understood. In our study, we present a new mechanism through which PLEXIND1 mediates its roles in cancer. For the first time, we demonstrate that it can function as a transforming growth factor beta coreceptor and modulate SMAD3 signaling. Around 90% of pancreatic cancer patients have mutant KRAS. Our work suggests that PLEXIND1 functions differently in pancreatic cancer cell lines, and the difference correlates with KRAS mutational status. Additionally, we demonstrate a novel peptide based therapeutic approach to target PLEXIND1 in cancer cells. Our work is valuable to both neuroscience and cancer fields, as it demonstrates an association between two previously unrelated signaling pathways. Abstract PLEXIND1 is upregulated in several cancers, including pancreatic ductal adenocarcinoma (PDAC). It is an established mediator of semaphorin signaling, and neuropilins are its known coreceptors. Herein, we report data to support the proposal that PLEXIND1 acts as a transforming growth factor beta (TGFβ) coreceptor, modulating cell growth through SMAD3 signaling. Our findings demonstrate that PLEXIND1 plays a pro-tumorigenic role in PDAC cells with oncogenic KRAS (KRASmut). We show in KRASmut PDAC cell lines (PANC-1, AsPC-1,4535) PLEXIND1 downregulation results in decreased cell viability (in vitro) and reduced tumor growth (in vivo). Conversely, PLEXIND1 acts as a tumor suppressor in the PDAC cell line (BxPC-3) with wild-type KRAS (KRASwt), as its reduced expression results in higher cell viability (in-vitro) and tumor growth (in vivo). Additionally, we demonstrate that PLEXIND1-mediated interactions can be selectively disrupted using a peptide based on its C-terminal sequence (a PDZ domain-binding motif), an outcome that may possess significant therapeutic implications. To our knowledge, this is the first report showing that (1) PLEXIND1 acts as a TGFβ coreceptor and mediates SMAD3 signaling, and (2) differential roles of PLEXIND1 in PDAC cell lines correlate with KRASmut and KRASwt status.
Collapse
Affiliation(s)
- Sneha Vivekanandhan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Vijay S. Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Thomas Caulfield
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Alexandre A. Pletnev
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
| | - Rosanna Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Yan W. Asmann
- Health Sciences Research, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - David Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Mark R. Spaller
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
- Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, NH 03756, USA
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan 215316, China
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
- Correspondence:
| |
Collapse
|
31
|
Timmerman DM, Remmers TL, Hillenius S, Looijenga LHJ. Mechanisms of TP53 Pathway Inactivation in Embryonic and Somatic Cells-Relevance for Understanding (Germ Cell) Tumorigenesis. Int J Mol Sci 2021; 22:ijms22105377. [PMID: 34065345 PMCID: PMC8161298 DOI: 10.3390/ijms22105377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 01/10/2023] Open
Abstract
The P53 pathway is the most important cellular pathway to maintain genomic and cellular integrity, both in embryonic and non-embryonic cells. Stress signals induce its activation, initiating autophagy or cell cycle arrest to enable DNA repair. The persistence of these signals causes either senescence or apoptosis. Over 50% of all solid tumors harbor mutations in TP53 that inactivate the pathway. The remaining cancers are suggested to harbor mutations in genes that regulate the P53 pathway such as its inhibitors Mouse Double Minute 2 and 4 (MDM2 and MDM4, respectively). Many reviews have already been dedicated to P53, MDM2, and MDM4, while this review additionally focuses on the other factors that can deregulate P53 signaling. We discuss that P14ARF (ARF) functions as a negative regulator of MDM2, explaining the frequent loss of ARF detected in cancers. The long non-coding RNA Antisense Non-coding RNA in the INK4 Locus (ANRIL) is encoded on the same locus as ARF, inhibiting ARF expression, thus contributing to the process of tumorigenesis. Mutations in tripartite motif (TRIM) proteins deregulate P53 signaling through their ubiquitin ligase activity. Several microRNAs (miRNAs) inactivate the P53 pathway through inhibition of translation. CCCTC-binding factor (CTCF) maintains an open chromatin structure at the TP53 locus, explaining its inactivation of CTCF during tumorigenesis. P21, a downstream effector of P53, has been found to be deregulated in different tumor types. This review provides a comprehensive overview of these factors that are known to deregulate the P53 pathway in both somatic and embryonic cells, as well as their malignant counterparts (i.e., somatic and germ cell tumors). It provides insights into which aspects still need to be unraveled to grasp their contribution to tumorigenesis, putatively leading to novel targets for effective cancer therapies.
Collapse
|
32
|
Zhu Z, Liu Z, Cui J, Huang Y, Chen H, Wu Y, Huang X, Gan C. Apoptosis inducing properties of 3-biotinylate-6-benzimidazole B-nor-cholesterol analogues. Steroids 2021; 169:108822. [PMID: 33722574 DOI: 10.1016/j.steroids.2021.108822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/14/2023]
Abstract
In this work, a series of Biotin-substituted B-nor-cholesteryl benzimidazole compounds were synthesized. The antiproliferativeactivities of these compounds were evaluated in vitro using a series of human cancer cell lines, including HeLa (cervical cancer), SKOV3 (ovarian cancer), T-47D (thymus gland cancer), MCF-7 (human breast cancer) and HEK293T (normal renal epithelial) cells. These compounds displayed distinct antiproliferative activities against the currently tested cancer cells. The apoptotic properties induced by compound 6d were further investigated. Our results showed that compound 6d could induce the apoptosis of SKOV3 cells, blocking the cell growth in S-phase. Western blotting analyses revealed that compound 6d can induce cell apoptosis via the mitochondria-dependent pathway.
Collapse
Affiliation(s)
- Zhiling Zhu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Zhiping Liu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Jianguo Cui
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yanmin Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Hualong Chen
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Yulan Wu
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Xiaotong Huang
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China
| | - Chunfang Gan
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, School of Chemistry and Material, Nanning Normal University, Nanning 530001, PR China.
| |
Collapse
|
33
|
Peng Y, Feng H, Wang C, Song Z, Zhang Y, Liu K, Cheng X, Zhao R. The role of E26 transformation-specific variant transcription factor 5 in colorectal cancer cell proliferation and cell cycle progression. Cell Death Dis 2021; 12:427. [PMID: 33931578 PMCID: PMC8087822 DOI: 10.1038/s41419-021-03717-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/03/2023]
Abstract
E26 transformation-specific variant transcription factor 5 (ETV5) contributes to tumor growth and progression and promotes colorectal cancer (CRC) angiogenesis. Previous studies indicate that ETV5 may regulate the cell cycle, but its detailed mechanism remain unclear. Gene Ontology (GO) analysis of RNA-seq data revealed that ETV5 possibly regulates the cell cycle in CRC. Here, in vitro and in vivo experiments were performed to verify that ETV5 promoted tumor progression and influenced cell cycle G1/S transition. Cell cycle PCR array and co-immunoprecipitation (Co-IP) helped identify the p21-CDKs pathway. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays were performed to determine whether ETV5 binds to the p21 promoter. ETV5 and p21 were detected by immunohistochemistry, and the effects of their expression on CRC patients were evaluated. ETV5 upregulation enhanced tumor proliferative capacity and promoted G1 phase transfer to the S phase. ETV5 knockdown slowed the growth of CRC cells and repressed the G1/S transition. We also found p21 as a downstream target of ETV5. p21 knockdown resulted in faster CRC cell growth and in more cells being driven from the G0/1 phase into the S phase. Co-IP experiments showed that p21 banding to CDK2, CDK4, and CDK6 inhibited p130 phosphorylation. Using the ChIP and luciferase reporter assay, we confirmed that ETV5 bound to the p21 promoter and repressed p21 expression. CRC patients with high ETV5 expression and low p21 expression showed the worst prognosis. Finally, by targeting p21 to regulate CDK function, ETV5 also changed drug-sensitivity to palbociclib and dinaciclib. In conclusion, ETV5 promoted cell cycle G1/S transition through transcriptional inhibition of p21, thereby accelerating tumor growth. Moreover, ETV5 changed drug-sensitivity to palbociclib and dinaciclib. Therefore, therapeutic regimens targeting ETV5 may be promising in improving the efficacy of target-CDK treatment in CRC.
Collapse
Affiliation(s)
- Yi Peng
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Haoran Feng
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Changgang Wang
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Zijia Song
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Yaqi Zhang
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Kun Liu
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Xi Cheng
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Ren Zhao
- grid.16821.3c0000 0004 0368 8293Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| |
Collapse
|
34
|
Ungerleider K, Beck J, Lissa D, Turnquist C, Horikawa I, Harris BT, Harris CC. Astrocyte senescence and SASP in neurodegeneration: tau joins the loop. Cell Cycle 2021; 20:752-764. [PMID: 33818291 DOI: 10.1080/15384101.2021.1909260] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tau accumulation is a core component of Alzheimer's disease and other neurodegenerative tauopathies. While tau's impact on neurons is a major area of research, the effect of extracellular tau on astrocytes is largely unknown. This article summarizes our recent studies showing that astrocyte senescence plays a critical role in neurodegenerative diseases and integrates extracellular tau into the regulatory loop of senescent astrocyte-mediated neurotoxicity. Human astrocytes in vitro undergoing senescence were shown to acquire the inflammatory senescence-associated secretory phenotype (SASP) and toxicity to neurons, which may recapitulate aging- and disease-associated neurodegeneration. Here, we show that human astrocytes exposed to extracellular tau in vitro also undergo cellular senescence and acquire a neurotoxic SASP (e.g. IL-6 secretion), with oxidative stress response (indicated by upregulated NRF2 target genes) and a possible activation of inflammasome (indicated by upregulated ASC and IL-1β). These findings suggest that senescent astrocytes induced by various conditions and insults, including tau exposure, may represent a therapeutic target to inhibit or delay the progression of neurodegenerative diseases. We also discuss the pathological activity of extracellular tau in microglia and astrocytes, the disease relevance and diversity of tau forms, therapeutics targeting senescence in neurodegeneration, and the roles of p53 and its isoforms in astrocyte-mediated neurotoxicity and neuroprotection.
Collapse
Affiliation(s)
- Kyra Ungerleider
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Comparative Pathobiology, Purdue University, West Layfette, Indiana, USA
| | - Delphine Lissa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Casmir Turnquist
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,University of Oxford Medical School, John Radcliffe Hospital, Oxford, UK
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brent T Harris
- Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Amani J, Gorjizadeh N, Younesi S, Najafi M, Ashrafi AM, Irian S, Gorjizadeh N, Azizian K. Cyclin-dependent kinase inhibitors (CDKIs) and the DNA damage response: The link between signaling pathways and cancer. DNA Repair (Amst) 2021; 102:103103. [PMID: 33812232 DOI: 10.1016/j.dnarep.2021.103103] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/16/2021] [Indexed: 02/08/2023]
Abstract
At the cellular level, DNA repair mechanisms are crucial in maintaining both genomic integrity and stability. DNA damage appears to be a central culprit in tumor onset and progression. Cyclin-dependent kinases (CDKs) and their regulatory partners coordinate the cell cycle progression. Aberrant CDK activity has been linked to a variety of cancers through deregulation of cell-cycle control. Besides DNA damaging agents and chromosome instability (CIN), disruptions in the levels of cell cycle regulators including cyclin-dependent kinase inhibitors (CDKIs) would result in unscheduled proliferation and cell division. The INK4 and Cip/Kip (CDK interacting protein/kinase inhibitor protein) family of CDKI proteins are involved in cell cycle regulation, transcription regulation, apoptosis, and cell migration. A thorough understanding of how these CDKIs regulate the DNA damage response through multiple signaling pathways may provide an opportunity to design efficient treatment strategies to inhibit carcinogenesis.
Collapse
Affiliation(s)
- Jafar Amani
- Applied Microbiology Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nassim Gorjizadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Simin Younesi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Mojtaba Najafi
- Department of Genetics, Faculty of Animal Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Golestan, Iran
| | - Arash M Ashrafi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Saeed Irian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Negar Gorjizadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Khalil Azizian
- Department of Clinical Microbiology, Sirjan School of Medical Sciences, Sirjan, Iran.
| |
Collapse
|
36
|
Tsitsipatis D, Grammatikakis I, Driscoll RK, Yang X, Abdelmohsen K, Harris SC, Yang JH, Herman AB, Chang MW, Munk R, Martindale JL, Mazan-Mamczarz K, De S, Lal A, Gorospe M. AUF1 ligand circPCNX reduces cell proliferation by competing with p21 mRNA to increase p21 production. Nucleic Acids Res 2021; 49:1631-1646. [PMID: 33444453 PMCID: PMC7897478 DOI: 10.1093/nar/gkaa1246] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/28/2020] [Accepted: 12/11/2020] [Indexed: 01/06/2023] Open
Abstract
Mammalian circRNAs can influence different cellular processes by interacting with proteins and other nucleic acids. Here, we used ribonucleoprotein immunoprecipitation (RIP) analysis to identify systematically the circRNAs associated with the cancer-related protein AUF1. Among the circRNAs interacting with AUF1 in HeLa (human cervical carcinoma) cells, we focused on hsa_circ_0032434 (circPCNX), an abundant target of AUF1. Overexpression of circPCNX specifically interfered with the binding of AUF1 to p21 (CDKN1A) mRNA, thereby promoting p21 mRNA stability and elevating the production of p21, a major inhibitor of cell proliferation. Conversely, silencing circPCNX increased AUF1 binding to p21 mRNA, reducing p21 production and promoting cell division. Importantly, eliminating the AUF1-binding region of circPCNX abrogated the rise in p21 levels and rescued proliferation. Therefore, we propose that the interaction of circPCNX with AUF1 selectively prevents AUF1 binding to p21 mRNA, leading to enhanced p21 mRNA stability and p21 protein production, thereby suppressing cell growth.
Collapse
Affiliation(s)
- Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Ioannis Grammatikakis
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute IRP, NIH, Bethesda, MD, USA
| | - Riley K Driscoll
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Xiaoling Yang
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Sophia C Harris
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jen-Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Allison B Herman
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Ming-Wen Chang
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Ashish Lal
- Regulatory RNAs and Cancer Section, Genetics Branch, Center for Cancer Research, National Cancer Institute IRP, NIH, Bethesda, MD, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| |
Collapse
|
37
|
Zhang HT, Gui T, Liu RX, Tong KL, Wu CJ, Li Z, Huang X, Xu QT, Yang J, Tang W, Sang Y, Liu W, Liu N, Ross RD, He QY, Zha ZG. Sequential targeting of YAP1 and p21 enhances the elimination of senescent cells induced by the BET inhibitor JQ1. Cell Death Dis 2021; 12:121. [PMID: 33495462 PMCID: PMC7835383 DOI: 10.1038/s41419-021-03416-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022]
Abstract
Chondrosarcoma (CHS) is the second most common bone malignancy with limited therapeutic approaches. Our previous study has found that Yes associated protein 1 (YAP1) is downregulated in CHS cells treated with bromodomain and extraterminal domain (BET) inhibitor JQ1. However, the precise role of YAP1 in CHS is largely unknown. Herein, we found that YAP1 expression was upregulated in CHS tissues, and positively correlated with its grading score. Loss of YAP1 inhibited CHS proliferation and induced cellular senescence, while expression of YAP1 mutants revealed YAP1/TEA domain family member (TEAD)-dependent negative regulation of p21 and subsequent cellular senescence. These results were validated by in vivo experiments using stable shYAP1 cell lines. Mechanistically, negative regulation of p21 by YAP1 occurred post-transcriptionally via Dicer-regulated miRNA networks, specifically, the miR-17 family. Furthermore, we demonstrated that sequential targeting of YAP1 and p21 enhanced the elimination of JQ1-induced senescent cells in a Bcl-2-like 1 (Bcl-XL)/Caspase-3 dependent manner. Altogether, we unveil a novel role of YAP1 signaling in mediating CHS cell senescence and propose a one-two punch approach that sequentially targets the YAP1/p21 axis to eliminate senescent cells.
Collapse
Affiliation(s)
- Huan-Tian Zhang
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China.
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China.
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Tao Gui
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ri-Xu Liu
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Kui-Leung Tong
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chong-Jie Wu
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhenyan Li
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xun Huang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Qiu-Tong Xu
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jie Yang
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wang Tang
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yuan Sang
- Department of Joint Replacement and Trauma Surgery, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wanting Liu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ning Liu
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ryan D Ross
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Zhen-Gang Zha
- Institute of Orthopedic Diseases, Jinan University, Guangzhou, China.
- Center for Joint Surgery and Sports Medicine, the First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
38
|
Wanner E, Thoppil H, Riabowol K. Senescence and Apoptosis: Architects of Mammalian Development. Front Cell Dev Biol 2021; 8:620089. [PMID: 33537310 PMCID: PMC7848110 DOI: 10.3389/fcell.2020.620089] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian development involves an exquisite choreography of cell division, differentiation, locomotion, programmed cell death, and senescence that directs the transformation of a single cell zygote to a mature organism containing on the order of 40 trillion cells in humans. How a single totipotent zygote undergoes the rapid stages of embryonic development to form over 200 different cell types is complex in the extreme and remains the focus of active research. Processes such as programmed cell death or apoptosis has long been known to occur during development to help sculpt organs and tissue systems. Other processes such as cellular senescence, long thought to only occur in pathologic states such as aging and tumorigenesis have been recently reported to play a vital role in development. In this review, we focus on apoptosis and senescence; the former as an integral mechanism that plays a critical role not only in mature organisms, but that is also essential in shaping mammalian development. The latter as a well-defined feature of aging for which some reports indicate a function in development. We will dissect the dual roles of major gene families, pathways such as Hox, Rb, p53, and epigenetic regulators such as the ING proteins in both early and the late stages and how they play antagonistic roles by increasing fitness and decreasing mortality early in life but contribute to deleterious effects and pathologies later in life.
Collapse
Affiliation(s)
- Emma Wanner
- Department of Biology, Faculty of Science, University of Calgary, Calgary, AB, Canada
| | - Harikrishnan Thoppil
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
39
|
Niu L, Yang S, Zhao X, Liu X, Si L, Wei M, Liu L, Cheng L, Qiao Y, Chen Z. Sericin inhibits MDA‑MB‑468 cell proliferation via the PI3K/Akt pathway in triple‑negative breast cancer. Mol Med Rep 2020; 23:140. [PMID: 33313947 PMCID: PMC7751468 DOI: 10.3892/mmr.2020.11779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/25/2020] [Indexed: 11/28/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer characterized by an aggressive histology and poor prognosis, with limited treatment options in the clinic. In the present study, the effect of sericin, as an anti-cancer drug, on TNBC cell proliferation was investigated using a MTT assay, a colony formation assay and immunocytochemistry staining of Ki67. Results from the flow cytometry demonstrated that sericin induced G0/G1 cell cycle arrest and promoted cellular apoptosis. Cell cycle and apoptosis-related proteins were detected via western blot analysis. Immunocytochemistry staining identified that P21 was translocated into the nucleus. Additionally, several pathways were significantly enriched in TNBC based on the Gene Expression Omnibus database, with the most prominent pathway being the PI3K/Akt signaling pathway. In TNBC MDA-MB-468 cells, sericin suppressed the PI3K/Akt pathway. All these findings suggested that sericin served a critical role in suppressing TNBC cell proliferation, inducing cell cycle arrest and promoting cellular apoptosis. The results indicated that the underlying molecular mechanism was, at least partially, via the downregulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Lin Niu
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Songhe Yang
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Xueying Zhao
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Xiaochao Liu
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lina Si
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Meng Wei
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lei Liu
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Luyang Cheng
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Yuebing Qiao
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhihong Chen
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
40
|
Exploring the extensive crosstalk between the antagonistic cytokines- TGF-β and TNF-α in regulating cancer pathogenesis. Cytokine 2020; 138:155348. [PMID: 33153895 DOI: 10.1016/j.cyto.2020.155348] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
A plethora of cytokines are produced in the tumor microenvironment (TME) those play a vital role in cancer prognosis. Though it is completely contextual, cytokines produced from an inflammatory micro-environment can either modulate cancer progression at early stages of tumor development or in later stages cytokine derived cues can in turn control tumor cell invasion and metastasis. Therefore, understanding the crosstalk between the key cytokines regulating cancer prognosis is critical for the development of an effective therapy. In this regard, the role of transforming growth factor-beta (TGF-β) in cancer is controversially discussed in general inhibition of TGF-β promotes de novo tumorigenesis whereas paradoxically, TGF-β can promote malignancy in already established tumors. Another important cytokine, TNF-α have intense crosstalk with TGF-β from the fact that in a non-cancer context, TGF-β promotes fibrosis whereas TNF-α has anti-fibrotic activity. We have recently reported that TGF-β-induced differentiation of epithelial cells to mesenchymal type is suppressed by TNF-α through regulation of cellular homeostatic machinery- autophagy. Moreover, there are also rare reports of synergy between these two cytokines as well. The crosstalk between TGF-β and TNF-α is not only limited to regulating cancer cell differentiation and proliferation but also includes involvement in cell death. In this review, we hence summarize the molecular mechanisms by which these two important cytokines, TGF-β and TNF-α control cancer prognosis.
Collapse
|
41
|
Swer PB, Sharma R. ATP-dependent chromatin remodelers in ageing and age-related disorders. Biogerontology 2020; 22:1-17. [PMID: 32968929 DOI: 10.1007/s10522-020-09899-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 11/27/2022]
Abstract
Ageing is characterized by the perturbation in cellular homeostasis associated with genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intracellular communication. Changes in the epigenome represent one of the crucial mechanisms during ageing and in age-related disorders. The ATP-dependent chromatin remodelers are an evolutionarily conserved family of nucleosome remodelling factors and generally regulate DNA repair, replication, recombination, transcription and cell cycle. Here, we review the chromatin based epigenetic changes that occur in ageing and age-related disorders with a specific reference to chromatin remodelers. We also discuss the link between dietary restriction and chromatin remodelers in regulating age-related processes with a view for consideration in future intervention studies.
Collapse
Affiliation(s)
- Pynskhem Bok Swer
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India.
| |
Collapse
|
42
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
43
|
Tsamou M, Nawrot TS, Carollo RM, Trippas AJ, Lefebvre W, Vanpoucke C, Vrijens K. Prenatal particulate air pollution exposure and expression of the miR-17/92 cluster in cord blood: Findings from the ENVIRONAGE birth cohort. ENVIRONMENT INTERNATIONAL 2020; 142:105860. [PMID: 32599355 DOI: 10.1016/j.envint.2020.105860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 05/28/2023]
Abstract
BACKGROUND Air pollution exposure during pregnancy is an important environmental health issue. Epigenetics mediate the effects of prenatal exposure and could increase disease predisposition in later life. The oncogenic miR-17/92 cluster is involved in normal development and disease. OBJECTIVES Here, for the first time the potential prenatal effects of particulate matter with a diameter<2.5 μm (PM2.5) exposure on expression of the miR-17/92 cluster in cord blood are explored. METHODS In 370 mother-newborn pairs from the ENVIRONAGE birth cohort, expression of three members of the miR-17/92 cluster was measured in cord blood by qRT-PCR. Expression of C-MYC and CDKN1A, a cluster activator and a target gene, respectively, was also analyzed. Multivariable linear regression models were used to associate the relative m(i)RNA expression with prenatal PM2.5 exposure. RESULTS PM2.5 exposure averaged (10th-90th percentile) 11.7 (9.0-14.4) µg/m3 over the entire pregnancy. In cord blood, miR-17 and miR-20a showed a -45.0% (95%CI: -55.9 to -31.4, p < 0.0001) and a -33.7% (95%CI: -46.9 to -17.2, p = 0.0003), decrease in expression in association with first trimester PM2.5 exposure, and a -32.5% (95%CI: -45.6 to -16.3, p = 0.0004) and -23.3% (95%CI: -38.1 to -4.8, p = 0.02), respectively, decrease in expression in association with PM2.5 exposure during the entire pregnancy. In association with third trimester PM2.5 exposure, a reduction of -25.8% (95%CI: -40.2 to -8.0, p = 0.007) and -14.2% (95%CI: -27.7 to 1.9, p = 0.08), for miR-20a and miR-92a expression, respectively, was identified. Only miR-92a expression (-15.7%, 95%CI: -27.3 to -2.4, p = 0.02) was associated with PM2.5 exposure during the last month of pregnancy. C-MYC expression was downregulated in cord blood in association with prenatal PM2.5 exposure during the first trimester and the entire pregnancy, in the adjusted model. DISCUSSION Lower expression levels of the miR-17/92 cluster in cord blood in association with increased prenatal PM2.5 exposure were observed. Whether this oncogenic microRNA cluster plays a role in trans-placental carcinogenesis remains to be elucidated.
Collapse
Affiliation(s)
- Maria Tsamou
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Tim S Nawrot
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium; Department of Public Health, Environment & Health Unit, Leuven University (KU Leuven), Leuven, Belgium
| | | | - Ann-Julie Trippas
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Wouter Lefebvre
- Flemish Institute for Technological Research (VITO), Mol, Belgium
| | | | - Karen Vrijens
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
44
|
Ma Y, Song Y, Shen R, Li P, Ding H, Guo Z, Liu X, Wang D. Loss of RAD6B induces degeneration of the cochlea in mice. Biochem Biophys Res Commun 2020; 531:402-408. [PMID: 32868078 DOI: 10.1016/j.bbrc.2020.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 01/11/2023]
Abstract
Presbycusis is a form of age-related hearing loss (AHL). Many studies have shown that the degeneration of various structures in the cochlea of the inner ear is related to AHL, and DNA damage is an important factor leading to the above process. As an E2 ubiquitin-conjugated enzyme, RAD6B plays an important role in DNA damage repair (DDR) through histone ubiquitination. However, the molecular mechanism is still unclear. In this study, we investigated the role of RAD6B in the morphological changes and DDR mechanisms in aging-related degeneration of the cochlea of mice. We observed that the hair cells, stria vascularis and spiral ganglion in the cochlea of the RAD6B knockout mice showed significant degenerative changes and abnormal expression of proteins associated with DDR mechanisms compared with those of the littermate wild-type mice. In conclusion, our results suggest that the deletion of RAD6B may lead to abnormalities in DDR, thereby accelerating the degeneration of various structures in the cochlea and senescence and apoptosis of cochlea cells.
Collapse
Affiliation(s)
- Yangping Ma
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yanfeng Song
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Rong Shen
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Panpan Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Han Ding
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Zhao Guo
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiangwen Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Degui Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
45
|
Mansilla SF, De La Vega MB, Calzetta NL, Siri SO, Gottifredi V. CDK-Independent and PCNA-Dependent Functions of p21 in DNA Replication. Genes (Basel) 2020; 11:genes11060593. [PMID: 32481484 PMCID: PMC7349641 DOI: 10.3390/genes11060593] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
p21Waf/CIP1 is a small unstructured protein that binds and inactivates cyclin-dependent kinases (CDKs). To this end, p21 levels increase following the activation of the p53 tumor suppressor. CDK inhibition by p21 triggers cell-cycle arrest in the G1 and G2 phases of the cell cycle. In the absence of exogenous insults causing replication stress, only residual p21 levels are prevalent that are insufficient to inhibit CDKs. However, research from different laboratories has demonstrated that these residual p21 levels in the S phase control DNA replication speed and origin firing to preserve genomic stability. Such an S-phase function of p21 depends fully on its ability to displace partners from chromatin-bound proliferating cell nuclear antigen (PCNA). Vice versa, PCNA also regulates p21 by preventing its upregulation in the S phase, even in the context of robust p21 induction by irradiation. Such a tight regulation of p21 in the S phase unveils the potential that CDK-independent functions of p21 may have for the improvement of cancer treatments.
Collapse
|
46
|
Hu Z, Long T, Ma Y, Zhu J, Gao L, Zhong Y, Wang X, Wang X, Li Z. Downregulation of GLYR1 contributes to microsatellite instability colorectal cancer by targeting p21 via the p38MAPK and PI3K/AKT pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:76. [PMID: 32370786 PMCID: PMC7201645 DOI: 10.1186/s13046-020-01578-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND GLYR1 has a high mutation frequency in microsatellite instability colorectal cancer (MSI CRC) and is presumed to be a novel tumor suppressor. However, the role of GLYR1 in tumors has never been studied. In particular, the downregulation of GLYR1 in MSI CRC is worthy of further investigation. METHODS Western blot and immunohistochemistry analyses were used to detect GLYR1 protein expression in CRC tissues and cell lines, and the clinical significance of GLYR1 was also analyzed. The relationship between GLYR1 and MLH1 was validated by immunofluorescence, immunoprecipitation and bioinformatics analyses. Western blotting, qRT-PCR, CCK-8 assays, colony formation assays, flow cytometry and Hoechst 33258 staining assays were used to assess the effect of GLYR1 on the cell cycle progression, proliferation, differentiation and apoptosis of CRC cells in vitro. The related mechanisms were initially investigated by Western blotting. RESULTS GLYR1 was significantly downregulated in MSI CRC and its expression was negatively correlated with tumor size and positively correlated with tumor differentiation in CRC patients. In addition, GLYR1 interacted with MLH1 to regulate its nuclear import and expression. Moreover, downregulation of GLYR1 accelerated G1/S phase transition, promoted proliferation and inhibited differentiation of SW480 and SW620 cells in vitro. Furthermore, downregulation of GLYR1 decreased the sensitivity to 5-fluorouracil (5-FU) by inhibiting the mitochondrial apoptosis pathway in CRC cells. Inhibition of the p38 mitogen-activated protein kinase (p38MAPK) and activation of the phosphatidyl 3-kinase/protein kinase B (PI3K/Akt) signaling pathways were involved in the mechanism by which GLYR1 downregulated p21. CONCLUSIONS Ours is the first study to elucidate the role of GLYR1 in tumors and provide evidence for GLYR1 as a biological marker that reflects the degree of malignancy and sensitivity to 5-FU in MSI CRC.
Collapse
Affiliation(s)
- Zhiyan Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Ting Long
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Yidan Ma
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Jiaxian Zhu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Lingfang Gao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Yan Zhong
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Xia Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Xiaoyan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China
| | - Zuguo Li
- Department of Pathology, Shenzhen Hospital of Southern Medical University, Shenzhen, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Molecular tumor Pathology, Guangzhou, China.
| |
Collapse
|
47
|
Heidari Z, Harati‐Sadegh M, Arian A, Maruei‐Milan R, Salimi S. The effect of
TP53
and
P21
gene polymorphisms on papillary thyroid carcinoma susceptibility and clinical/pathological features. IUBMB Life 2020; 72:922-930. [DOI: 10.1002/iub.2225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Zahra Heidari
- Department of EndocrinologySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Mahdiyeh Harati‐Sadegh
- Genetic of Non‐Communicable Disease Research CenterZahedan University of Medical Sciences Zahedan Iran
| | - Abtin Arian
- Department of RadiologySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Rostam Maruei‐Milan
- Department of Clinical BiochemistrySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
| | - Saeedeh Salimi
- Department of Clinical BiochemistrySchool of Medicine, Zahedan University of Medical Sciences Zahedan Iran
- Cellular and Molecular Research Center, Resistant Tuberculosis InstituteZahedan University of Medical Sciences Zahedan Iran
| |
Collapse
|
48
|
Loss of CDKN1A mRNA and Protein Expression Are Independent Predictors of Poor Outcome in Chromophobe Renal Cell Carcinoma Patients. Cancers (Basel) 2020; 12:cancers12020465. [PMID: 32079343 PMCID: PMC7072616 DOI: 10.3390/cancers12020465] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/24/2020] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
Chromophobe renal cell carcinoma (chRCC) patients have good prognosis. Only 5%-10% patients die of metastatic disease after tumorectomy, but tumor progression cannot be predicted by histopathological parameters alone. chRCC are characterized by losses of many chromosomes, whereas gene mutations are rare. In this study, we aim at identifying genes indicating chRCC progression. A bioinformatic approach was used to correlate chromosomal loss and mRNA expression from 15287 genes from The Cancer Genome Atlas (TCGA) database. All genes in TCGA chromophobe renal cancer dataset (KICH) for which a significant correlation between chromosomal loss and mRNA expression was shown, were identified and their associations with outcome was assessed. Genome-wide DNA copy-number alterations were analyzed by Affymetrix OncoScan® CNV FFPE Microarrays in a second cohort of Swiss chRCC. In both cohorts, tumors with loss of chromosomes 2, 6, 10, 13, 17 and 21 had signs of tumor progression. There were 4654 genes located on these chromosomes, and 13 of these genes had reduced mRNA levels, which was associated with poor outcome in chRCC. Decreased CDKN1A expression at mRNA (p = 0.02) and protein levels (p = 0.02) were associated with short overall survival and were independent predictors of prognosis (p <0.01 and <0.05 respectively). CDKN1A expression status is a prognostic biomarker independent of tumor stage. CDKN1A immunohistochemistry may be used to identify chRCC patients at greater risk of disease progression.
Collapse
|
49
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
50
|
Senile Osteoporosis: The Involvement of Differentiation and Senescence of Bone Marrow Stromal Cells. Int J Mol Sci 2020; 21:ijms21010349. [PMID: 31948061 PMCID: PMC6981793 DOI: 10.3390/ijms21010349] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
Senile osteoporosis has become a worldwide bone disease with the aging of the world population. It increases the risk of bone fracture and seriously affects human health. Unlike postmenopausal osteoporosis which is linked to menopause in women, senile osteoporosis is due to aging, hence, affecting both men and women. It is commonly found in people with more than their 70s. Evidence has shown that with age increase, bone marrow stromal cells (BMSCs) differentiate into more adipocytes rather than osteoblasts and undergo senescence, which leads to decreased bone formation and contributes to senile osteoporosis. Therefore, it is necessary to uncover the molecular mechanisms underlying the functional changes of BMSCs. It will benefit not only for understanding the senile osteoporosis development, but also for finding new therapies to treat senile osteoporosis. Here, we review the recent advances of the functional alterations of BMSCs and the related mechanisms during senile osteoporosis development. Moreover, the treatment of senile osteoporosis by aiming at BMSCs is introduced.
Collapse
|