51
|
Zaborowska M, Vazirisani F, Shah FA, Firdaus R, Omar O, Ekström K, Trobos M, Thomsen P. Immunomodulatory effects exerted by extracellular vesicles from Staphylococcus epidermidis and Staphylococcus aureus isolated from bone-anchored prostheses. Biomaterials 2021; 278:121158. [PMID: 34619562 DOI: 10.1016/j.biomaterials.2021.121158] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/11/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are the bacteria that most frequently cause osteomyelitis. This study aimed to determine whether staphylococci isolated from osteomyelitis associated with septic loosening of orthopedic prostheses release extracellular vesicles (EVs) and, if so, to determine tentative immunomodulatory effects on the human monocytic cell line THP-1. EVs were isolated from bacterial cultures using filtration and ultracentrifugation and characterized by scanning electron microscopy, nanoparticle tracking analysis and Western Blot. The cytotoxic effect of EVs was analyzed by NucleoCounter and lactate dehydrogenase (LDH) analyses. Confocal laser scanning microscopy was employed to visualize the uptake of EVs by THP-1 cells. Activation of the transcription factor nuclear factor-κB (NF-κB) was determined in THP1-Blue™ NF-κB cells, and the gene expression and secretion of cytokines were determined by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. All investigated strains, irrespective of their biofilm formation ability, were able to secrete EVs in vitro. The S. aureus strains produced significantly more EVs than the S. epidermidis strains. Both S. aureus-derived EVs and S. epidermidis-derived EVs were internalized by THP-1 cells, upregulated Toll-like receptor 3 (TLR3) gene expression, activated NF-κB, and promoted the gene expression and secretion of interleukin (IL)-8, monocyte chemoattractant protein (MCP)-1, matrix metallopeptidase (MMP)-9 and IL-10. Whereas EVs from both staphylococcal species upregulated the proapoptotic DNA damage-inducible transcript 4 (DDIT4) gene and downregulated the antiapoptotic B-cell lymphoma 2 (Bcl-2) gene, cytolysis was preferentially induced in S. aureus EV-stimulated cells, possibly related to the expression of cytolytic proteins predominantly in S. aureus EVs. In conclusion, staphylococcal EVs possess potent cytolytic and immunomodulatory properties.
Collapse
Affiliation(s)
- Magdalena Zaborowska
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Forugh Vazirisani
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Furqan A Shah
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rininta Firdaus
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Ekström
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Center for Antibiotic Resistance Research (CARe), University of Gothenburg, Gothenburg, Sweden
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
52
|
Zhang D, Li Y, Yang S, Wang M, Yao J, Zheng Y, Deng Y, Li N, Wei B, Wu Y, Zhai Z, Dai Z, Kang H. Identification of a glycolysis-related gene signature for survival prediction of ovarian cancer patients. Cancer Med 2021; 10:8222-8237. [PMID: 34609082 PMCID: PMC8607265 DOI: 10.1002/cam4.4317] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 08/22/2021] [Accepted: 08/31/2021] [Indexed: 12/17/2022] Open
Abstract
Background Ovarian cancer (OV) is deemed the most lethal gynecological cancer in women. The aim of this study was to construct an effective gene prognostic model for predicting overall survival (OS) in patients with OV. Methods The expression profiles of glycolysis‐related genes (GRGs) and clinical data of patients with OV were extracted from The Cancer Genome Atlas (TCGA) database. Univariate, multivariate, and least absolute shrinkage and selection operator Cox regression analyses were conducted, and a prognostic signature based on GRGs was constructed. The predictive ability of the signature was analyzed using training and test sets. Results A gene risk signature based on nine GRGs (ISG20, CITED2, PYGB, IRS2, ANGPTL4, TGFBI, LHX9, PC, and DDIT4) was identified to predict the survival outcome of patients with OV. The signature showed a good prognostic ability for OV, particularly high‐grade OV, in the TCGA dataset, with areas under the curve (AUC) of 0.709 and 0.762 for 3‐ and 5‐year survival, respectively. Similar results were found in the test sets, and the AUCs of 3‐, 5‐year OS were 0.714 and 0.772 in the combined test set. And our signature was an independent prognostic factor. Moreover, a nomogram combining the prediction model and clinical factors was developed. Conclusion Our study established a nine‐GRG risk model and nomogram to better predict OS in patients with OV. The risk model represents a promising and independent prognostic predictor for patients with OV. Moreover, our study on GRGs could offer guidance for the elucidation of underlying mechanisms in future studies.
Collapse
Affiliation(s)
- Dai Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, China
| | - Yiche Li
- Department of Tumor Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Si Yang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Yao
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zheng
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yujiao Deng
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bajin Wei
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Zhai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
53
|
The REDD1/TXNIP Complex Accelerates Oxidative Stress-Induced Apoptosis of Nucleus Pulposus Cells through the Mitochondrial Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7397516. [PMID: 34603601 PMCID: PMC8481043 DOI: 10.1155/2021/7397516] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/07/2021] [Indexed: 01/19/2023]
Abstract
The death of nucleus pulposus (NP) cells is an important cause of intervertebral disc (IVD) degeneration. Redox disturbance caused by dysfunctional mitochondria has been considered as a vital risk for NP cell survival. It is valuable to identify key proteins maintaining mitochondrial function in NP cells. A previous study found that regulated in development and DNA damage response 1 (REDD1) are upregulated during intervertebral disc degeneration and that REDD1 can cause NP cell apoptosis. Thus, the present study further explores the effect of REDD1 on IVD degeneration. Our results showed that REDD1 promotes NP cell apoptosis via the mitochondrial pathway. Importantly, REDD1 formed a complex with TXNIP to strengthen its own action, and the combination was consolidated under H2O2-induced oxidative stress. The combined inhibition of the REDD1/TXNIP complex was better than that of REDD1 or TXNIP alone in restoring cell proliferation and accelerating apoptosis. Moreover, p53 acts as the transcription factor of REDD1 to regulate the REDD1/TXNIP complex under oxidative stress. Altogether, our results demonstrated that the REDD1/TXNIP complex mediated H2O2-induced human NP cell apoptosis and IVD degeneration through the mitochondrial pathway. Interferences on these sites to achieve mitochondrial redox homeostasis may be a novel therapeutic strategy for oxidative stress-associated IVD degeneration.
Collapse
|
54
|
Lau HC, Shen Y, Huang Q, Huang HY, Zhou L. Glycolysis related gene expression signature in predicting prognosis of laryngeal squamous cell carcinoma. Bioengineered 2021; 12:8738-8752. [PMID: 34565301 PMCID: PMC8806568 DOI: 10.1080/21655979.2021.1980177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Researches have suggested that aerobic glycolysis can reflect the development and progression of most carcinomas. We aimed to investigate whether glycolysis-related genes (GRGs) are associated with overall survival in laryngeal squamous cell carcinoma (LSCC). Here, we identified differentially expressed GRGs in TCGA dataset and microarray sample of GSE27020 from GEO database. A set of two glycolytic gene signatures, including DDIT4 and PLOD2 was screened through Cox and Lasso regression. The risk score was calculated using the gene expression of the two GRGs. The high-risk group presented a poor prognosis through Kaplan–Meier method. The ROC curve indicated good prediction performance in survival based on the validation of four cohorts. Univariate and multivariate Cox regression analyses suggested that two-gene signature could be an independent risk factor in LSCC. A total of 17 LSCC patients were enrolled to clarify the genetic expression through using reverse transcription-polymerase chain reaction (RT-PCR). A visualized nomogram was then constructed to predict 1-, 3-, and 5-year overall survival. Taken together, two novel glycolytic gene signatures were discovered and validated, providing a potential therapeutic and overall survival (OS)-prediction biomarker for LSCC.
Collapse
Affiliation(s)
- Hui-Ching Lau
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Clinical Disciplines of Otorhinolaryngology, Shanghai, PR China
| | - Yujie Shen
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Clinical Disciplines of Otorhinolaryngology, Shanghai, PR China
| | - Qiang Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Clinical Disciplines of Otorhinolaryngology, Shanghai, PR China
| | - Hui-Ying Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Clinical Disciplines of Otorhinolaryngology, Shanghai, PR China
| | - Liang Zhou
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,Shanghai Key Clinical Disciplines of Otorhinolaryngology, Shanghai, PR China
| |
Collapse
|
55
|
Potential Roles of Sestrin2 in Alzheimer's Disease: Antioxidation, Autophagy Promotion, and Beyond. Biomedicines 2021; 9:biomedicines9101308. [PMID: 34680426 PMCID: PMC8533411 DOI: 10.3390/biomedicines9101308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common age-related neurodegenerative disease. It presents with progressive memory loss, worsens cognitive functions to the point of disability, and causes heavy socioeconomic burdens to patients, their families, and society as a whole. The underlying pathogenic mechanisms of AD are complex and may involve excitotoxicity, excessive generation of reactive oxygen species (ROS), aberrant cell cycle reentry, impaired mitochondrial function, and DNA damage. Up to now, there is no effective treatment available for AD, and it is therefore urgent to develop an effective therapeutic regimen for this devastating disease. Sestrin2, belonging to the sestrin family, can counteract oxidative stress, reduce activity of the mammalian/mechanistic target of rapamycin (mTOR), and improve cell survival. It may therefore play a crucial role in neurodegenerative diseases like AD. However, only limited studies of sestrin2 and AD have been conducted up to now. In this article, we discuss current experimental evidence to demonstrate the potential roles of sestrin2 in treating neurodegenerative diseases, focusing specifically on AD. Strategies for augmenting sestrin2 expression may strengthen neurons, adapting them to stressful conditions through counteracting oxidative stress, and may also adjust the autophagy process, these two effects together conferring neuronal resistance in cases of AD.
Collapse
|
56
|
Feng Y, Cao X, Zhao B, Song C, Pang B, Hu L, Zhang C, Wang J, He J, Wang S. Nitrate increases cisplatin chemosensitivity of oral squamous cell carcinoma via REDD1/AKT signaling pathway. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1814-1828. [PMID: 34542810 DOI: 10.1007/s11427-020-1978-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
Although cisplatin is one of the chemotherapeutics most frequently used in oral squamous cell carcinoma (OSCC) treatment, it exerts multiple side effects and poor chemosensitivity. Nitrate reportedly demonstrates several beneficial biological functions, and synthesized nitrates enhance the therapeutic efficacy of chemotherapy. However, the role of inorganic nitrate in cisplatin chemotherapy remains unclear. We therefore investigated the effect of inorganic nitrate exerted on cisplatin sensitivity in OSCC. We found that nitrate did not affect OSCC cell growth and apoptosis in OSCC cells and OSCC xenograft tumor animal studies. Cisplatin induced REDD1 expression and AKT activation in OSCC. However, nitrate could increase cisplatin chemosensitivity, reduce its REDD1 expression, and attenuate AKT signaling activation in OSCC cells. Dysregulation of high levels of REDD1, which could enhance AKT activation, was positively associated with poor prognosis in OSCC patients. Thus, reduced REDD1 expression and retarded AKT activation induced by inorganic nitrate might be a new potential approach to the sensitization of oral cancer to cisplatin treatment in the future.
Collapse
Affiliation(s)
- Yuanyong Feng
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Xuedi Cao
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Bin Zhao
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Chunyan Song
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Baoxing Pang
- Department of Oral and Maxillofacial Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Liang Hu
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Chunmei Zhang
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Jinsong Wang
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China
| | - Junqi He
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China.
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, 100050, China.
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, Beijing, 100069, China.
- Research Units of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100069, China.
| |
Collapse
|
57
|
Song L, Chen Z, Zhang M, Zhang M, Lu X, Li C, Miao L. DDIT4 overexpression associates with poor prognosis in lung adenocarcinoma. J Cancer 2021; 12:6422-6428. [PMID: 34659532 PMCID: PMC8489140 DOI: 10.7150/jca.60118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/18/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives: DNA damage inducible transcript 4 (DDIT4) plays a key role in different cancers, but the role of DDIT4 in lung adenocarcinoma (LUAD) is not completely understood. The aim of this study was to evaluate the utility of DDIT4 as a prognostic biomarker for LUAD. Methods: First, DDIT4 mRNA expression in LUAD cell lines (A549, H1299 and HBE) and tissues (89 cases) was assessed by RT-PCR. Next, DDIT4 protein expression in LUAD tissues and normal tissues was assessed by immunohistochemistry (75 cases). Then, the correlation between DDIT4 expression and overall survival was analyzed using the Kaplan-Meier method. After that, we verified the utility of the DDIT4 gene as a prognostic marker of lung cancer in the TCGA database (1133 cases). Finally, the possible mechanism of the DDIT4 gene as a prognostic marker of LUAD was preliminarily explored. Results: mRNA levels of DDIT4 in HBE cells were significantly lower than in A549 and H1299 cells (P<0.05), and expression of the DDIT4 gene in cancer tissues was significantly higher than in adjacent tissues (P<0.0001). Immunohistochemical staining results showed that high expression of DDIT4 accounted for approximately 68.0% of LUAD tissues. DDIT4 expression was significantly correlated with differentiation (P < 0.05). However, it was not correlated with sex, age, smoking, tumor size, lymph node metastasis, or TNM stage (P>0.05). The survival analysis demonstrated that high DDIT4 expression was correlated with shorter overall survival (P < 0.05). Univariate and multivariate analyses indicated that DDIT4 was an independent predictor of overall survival for LUAD, which was confirmed by data from the TCGA database. Finally, we found that DDIT4 gene expression was significantly increased in the hypoxic environment compared to the normal oxygen environment, indicating that the DDIT4 gene may play an important role in the hypoxic microenvironment of tumor tissue. Conclusion: High expression levels of DDIT4 correlated with poor overall survival in patients with LUAD, and DDIT4 was an independent predictor of overall survival. These findings provide new insight for understanding the development of LUAD.
Collapse
Affiliation(s)
- Li Song
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China, 215006
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Zhiyao Chen
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China, 215006
| | - Menghua Zhang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China, 215006
| | - Mingli Zhang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China, 215006
| | - Xiaoyun Lu
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Chunsun Li
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China, 215006
| |
Collapse
|
58
|
Li Z, Zhao Q, Lu Y, Zhang Y, Li L, Li M, Chen X, Sun D, Duan Y, Xu Y. DDIT4 S-Nitrosylation Aids p38-MAPK Signaling Complex Assembly to Promote Hepatic Reactive Oxygen Species Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101957. [PMID: 34310076 PMCID: PMC8456271 DOI: 10.1002/advs.202101957] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/31/2021] [Indexed: 05/11/2023]
Abstract
Mitogen-activated protein kinase (MAPK) signaling plays a significant role in reactive oxygen species (ROS) production. The authors have previously shown that Brahma-related gene 1 (BRG1), a chromatin remodeling protein, contributes to hepatic ROS accumulation in multiple animal and cellular models of liver injury. Here it is reported that DNA damage-induced transcript 4 (DDIT4) is identified as a direct transcriptional target for BRG1. DDIT4 overexpression overcomes BRG1 deficiency to restore ROS production whereas DDIT4 knockdown phenocopies BRG1 deficiency in suppressing ROS production in vitro and in vivo. Mechanistically, DDIT4 coordinates the assembly of the p38-MAPK signaling complex to drive ROS production in an S-nitrosylation dependent manner. Molecular docking identifies several bioactive DDIT4-inteacting compounds including imatinib, nilotinib, and nateglinide, all of which are confirmed to attenuate hepatic ROS production, dampen p38-MAPK signaling, and ameliorate liver injury by influencing DDIT4 S-nitrosylation. Importantly, positive correlation between ROS levels and BRG1/DDIT4/S-nitrosylated DDIT4 levels is detected in human liver biopsy specimens. In conclusion, the data reveal a transcription-based signaling cascade that contributes to ROS production in liver injury.
Collapse
Affiliation(s)
- Zilong Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of ChangzhouThe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Translational MedicineNanjing Medical UniversityNanjing211166China
- Institute of Biomedical ResearchLiaocheng UniversityLiaocheng252000China
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Qianwen Zhao
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Yunjie Lu
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of ChangzhouThe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yangxi Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Translational MedicineNanjing Medical UniversityNanjing211166China
| | - Luyang Li
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Translational MedicineNanjing Medical UniversityNanjing211166China
| | - Min Li
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Translational MedicineNanjing Medical UniversityNanjing211166China
| | - Xuemin Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of ChangzhouThe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Donglin Sun
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of ChangzhouThe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yunfei Duan
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of ChangzhouThe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Translational MedicineNanjing Medical UniversityNanjing211166China
- Institute of Biomedical ResearchLiaocheng UniversityLiaocheng252000China
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
59
|
Kuznetsova AV, Rzhanova LA, Aleksandrova MA. Small Noncoding RNA in Regulation of Differentiation of Retinal Pigment Epithelium. Russ J Dev Biol 2021. [DOI: 10.1134/s106236042103005x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
60
|
Transcriptome sequencing provides insights into the mechanism of hypoxia adaption in bighead carp (Hypophthalmichthys nobilis). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100891. [PMID: 34404015 DOI: 10.1016/j.cbd.2021.100891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
Hypoxia negatively affects the behavior, immunology, physiology, and growth of fish. Therefore, uncovering the genetic mechanisms underlying hypoxia adaptation and tolerance in fish prior to any genetic improvement is essential. Bighead carp is one of the most important freshwater fish species in aquaculture worldwide; however, this species does not have a strong ability to tolerate hypoxia. In this study, the dissolved oxygen level (0.6 mg/L) was maintained above the asphyxiation point of bighead carp for a long time to simulate hypoxia stress. The liver, gills, and heart were sampled before (0 h) and after (1 h, 2 h, 4 h) the hypoxia tests. Glutathione peroxidase (GPx) and catalase (CAT) activities and malondialdehyde (MDA) levels in the liver were significantly (p < 0.05) elevated at 1 h after hypoxic stress. By observing tissue morphology, the cell structure of the liver and gill tissues was found to change to varying degrees before and after hypoxia stress. Transcriptome sequencing was performed on 36 samples of gill, liver, and heart at four time points, and a total of 293.55G of data was obtained. In the early phase (0-1 h), differentially expressed genes (DEGs, 807 genes upregulated, 654 genes downregulated) were mainly enriched in signal transduction, such as cytokine-cytokine receptor interactions and ECM-receptor interactions. In the middle phase (0-2 h), DEGs (1201 genes upregulated and 2036 genes downregulated) were mainly enriched in regulation and adaptation, such as the MAPK and FoxO signaling pathways. Finally, in the later phase (0-4 h), DEGs (3975 genes upregulated and 4412 genes downregulated) were mainly enriched in tolerance and apoptosis, such as the VEGF signaling pathway and apoptosis. The genes with the most remarkable upregulation at different time points in the three tissues had some similarities. Genetic differences in these genes may be responsible for the differences in hypoxia tolerance among individuals. Altogether, our study provides new insights into the molecular mechanisms of hypoxia adaptation in fish. Further, the key regulatory genes identified provide genetic resources for breeding hypoxia-tolerant bighead carp species.
Collapse
|
61
|
Masumoto S, Ono A, Ito A, Kawabe Y, Kamihira M. Hypoxia-responsive expression of vascular endothelial growth factor for induction of angiogenesis in artificial three-dimensional tissues. J Biosci Bioeng 2021; 132:399-407. [PMID: 34364783 DOI: 10.1016/j.jbiosc.2021.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022]
Abstract
Constructing three-dimensional (3D) tissues is an important process to improve cellular functions in tissue engineering. When transplanting artificially constructed tissues, a poor vascular network restricts oxygen and nutrient supplies to the tissue cells, which leads to cell death and reduced rates of tissue engraftment. Therefore, it is necessary to develop a system that builds a vascular network within 3D tissues. Here, we developed a hypoxia-responsive gene expression system for production of an angiogenic factor, vascular endothelial growth factor (VEGF), to improve hypoxia and nutrition deficiencies inside artificial 3D tissues. We demonstrated that cells into which the hypoxia-responsive VEGF gene expression system had been introduced autonomously controlled VEGF expression in a hypoxic stress-dependent manner. Next, we confirmed that VEGF expression within a 3D cell sheet was induced in response to a hypoxic environment in vitro. The genetically modified cell sheet was subcutaneously transplanted into mice to evaluate the feasibility of the hypoxia-responsive VEGF gene expression system in vivo. The results suggest that the hypoxia-responsive VEGF gene expression system is promising to prepare artificial 3D tissues in regenerative medicine.
Collapse
Affiliation(s)
- Shinya Masumoto
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akihiko Ono
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
62
|
Doan P, Nguyen P, Murugesan A, Subramanian K, Konda Mani S, Kalimuthu V, Abraham BG, Stringer BW, Balamuthu K, Yli-Harja O, Kandhavelu M. Targeting Orphan G Protein-Coupled Receptor 17 with T0 Ligand Impairs Glioblastoma Growth. Cancers (Basel) 2021; 13:cancers13153773. [PMID: 34359676 PMCID: PMC8345100 DOI: 10.3390/cancers13153773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/10/2021] [Accepted: 07/22/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM), or glioblastoma chemotherapy, has one of the poorest improvements across all types of cancers. Despite the different rationales explored in targeted therapy for taming the GBM aggressiveness, its phenotypic plasticity, drug toxicity, and adaptive resistance mechanisms pose many challenges in finding an effective cure. Our manuscript reports the expression and prognostic role of orphan receptor GPR17 in glioma, the molecular mechanism of action of the novel ligand of GPR17, and provides evidence how the T0 agonist promotes glioblastoma cell death through modulation of the MAPK/ERK, PI3K–Akt, STAT, and NF-κB pathways. The highlights are as follows: GPR17 expression is associated with greater survival for both low-grade glioma (LGG) and GBM; GA-T0, a potent GPR17 receptor agonist, causes significant GBM cell death and apoptosis; GPR17 signaling promotes cell cycle arrest at the G1 phase in GBM cells; key genes are modulated in the signaling pathways that inhibit GBM cell proliferation; and GA-T0 crosses the blood–brain barrier and reduces tumor volume. Abstract Glioblastoma, an invasive high-grade brain cancer, exhibits numerous treatment challenges. Amongst the current therapies, targeting functional receptors and active signaling pathways were found to be a potential approach for treating GBM. We exploited the role of endogenous expression of GPR17, a G protein-coupled receptor (GPCR), with agonist GA-T0 in the survival and treatment of GBM. RNA sequencing was performed to understand the association of GPR17 expression with LGG and GBM. RT-PCR and immunoblotting were performed to confirm the endogenous expression of GPR17 mRNA and its encoded protein. Biological functions of GPR17 in the GBM cells was assessed by in vitro analysis. HPLC and histopathology in wild mice and an acute-toxicity analysis in a patient-derived xenograft model were performed to understand the clinical implication of GA-T0 targeting GPR17. We observed the upregulation of GPR17 in association with improved survival of LGG and GBM, confirming it as a predictive biomarker. GA-T0-stimulated GPR17 leads to the inhibition of cyclic AMP and calcium flux. GPR17 signaling activation enhances cytotoxicity against GBM cells and, in patient tissue-derived mesenchymal subtype GBM cells, induces apoptosis and prevents proliferation by stoppage of the cell cycle at the G1 phase. Modulation of the key genes involved in DNA damage, cell cycle arrest, and in several signaling pathways, including MAPK/ERK, PI3K–Akt, STAT, and NF-κB, prevents tumor regression. In vivo activation of GPR17 by GA-T0 reduces the tumor volume, uncovering the potential of GA-T0–GPR17 as a targeted therapy for GBM treatment. Conclusion: Our analysis suggests that GA-T0 targeting the GPR17 receptor presents a novel therapy for treating glioblastoma.
Collapse
Affiliation(s)
- Phuong Doan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | - Phung Nguyen
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
- Department of Biotechnology, Lady Doak College, Thallakulam, Madurai 625002, India
| | - Kumar Subramanian
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
| | | | - Vignesh Kalimuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (K.B.)
| | - Bobin George Abraham
- Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland;
| | - Brett W. Stringer
- College of Medicine and Public Health, Flinders University, Sturt Rd., Bedford Park, SA 5042, Australia;
| | - Kadalmani Balamuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (K.B.)
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland;
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland; (P.D.); (P.N.); (A.M.); (K.S.)
- BioMediTech Institute and Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön Katu 34, 33520 Tampere, Finland
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
- Correspondence: ; Tel.: +358-504721724
| |
Collapse
|
63
|
Yi YW, Park NY, Park JI, Seong YS, Hong YB. Doxycycline potentiates the anti-proliferation effects of gemcitabine in pancreatic cancer cells. Am J Cancer Res 2021; 11:3515-3536. [PMID: 34354858 PMCID: PMC8332860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/13/2021] [Indexed: 06/13/2023] Open
Abstract
Gemcitabine is often recommended as a first-line treatment for patients with metastatic pancreatic cancer. However, gemcitabine resistance is a major challenge in the treatment of pancreatic ductal adenocarcinoma. Our group serendipitously identified the role of doxycycline as a potentiator of gemcitabine efficacy in pancreatic cancer cells. Doxycycline and gemcitabine co-treatment was significantly more cytotoxic to pancreatic cancer cells compared to gemcitabine alone. Interestingly, doxycycline only exerted synergistic effects when coupled with gemcitabine as opposed to other conventional chemotherapeutics including nucleoside analogs. The anti-clonogenic effects of gemcitabine on pancreatic cancer cells were also enhanced by doxycycline. According to cell cycle analyses, doxycycline prolonged gemcitabine-mediated S phase cell cycle arrest. Further, gene expression profiling analyses indicated that a small set of genes involved in cell cycle regulation were uniquely modulated by gemcitabine and doxycycline co-treatment compared to gemcitabine alone. Western blot analyses indicated that several cell cycle-related proteins, including cyclin D1, p21, and DNA damage inducible transcript 4 (DDIT4), were further modulated by doxycycline and gemcitabine co-treatment. Taken together, our findings indicate that doxycycline enhances the effects of gemcitabine on cell cycle progression, thus rendering pancreatic cancer cells more sensitive to gemcitabine. However, additional studies are required to assess the mechanisms of doxycycline and gemcitabine synergism, which might lead to novel treatment options for pancreatic cancer.
Collapse
Affiliation(s)
- Yong Weon Yi
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook UniversityCheonan, Korea
| | - Na Young Park
- Department of Translational Biomedical Sciences, Graduate School of Dong-A UniversityBusan 49201, Korea
| | - Joo-In Park
- Department of Translational Biomedical Sciences, Graduate School of Dong-A UniversityBusan 49201, Korea
- Department of Biochemistry, College of Medicine, Dong-A UniversityBusan 49201, Korea
| | - Yeon-Sun Seong
- Department of Nanobiomedical Science and BK21 PLUS Research Center for Regenerative Medicine, Dankook UniversityCheonan, Korea
- Department of Biochemistry, College of Medicine, Dankook UniversityCheonan 31116, Korea
- Graduate School of Convergence Medical Science, Dankook UniversityCheonan 31116, Korea
| | - Young Bin Hong
- Department of Translational Biomedical Sciences, Graduate School of Dong-A UniversityBusan 49201, Korea
- Department of Biochemistry, College of Medicine, Dong-A UniversityBusan 49201, Korea
| |
Collapse
|
64
|
Zamora-Briseño JA, Améndola-Pimenta M, Ortega-Rosas DA, Pereira-Santana A, Hernández-Velázquez IM, González-Penagos CE, Pérez-Vega JA, Del Río-García M, Árcega-Cabrera F, Rodríguez-Canul R. Gill and liver transcriptomic responses of Achirus lineatus (Neopterygii: Achiridae) exposed to water-accommodated fraction (WAF) of light crude oil reveal an onset of hypoxia-like condition. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34309-34327. [PMID: 33646544 DOI: 10.1007/s11356-021-12909-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
Crude oil is one of the most widespread pollutants released into the marine environment, and native species have provided useful information about the effect of crude oil pollution in marine ecosystems. We consider that the lined sole Achirus lineatus can be a useful monitor of the effect of crude oil in the Gulf of Mexico (GoM) because this flounder species has a wide distribution along the GoM, and its response to oil components is relevant. The objective of this study was to compare the transcriptomic changes in liver and gill of adults lined sole fish (Achirus lineatus) exposed to a sublethal acute concentration of water-accommodated fraction (WAF) of light crude oil for 48 h. RNA-Seq was performed to assess the transcriptional changes in both organs. A total of 1073 differentially expressed genes (DEGs) were detected in gills; 662 (61.69%) were upregulated, and 411 (38.30%) were downregulated whereas in liver, 515 DEGs; 306 (59.42%) were upregulated, and 209 (40.58%) were downregulated. Xenobiotic metabolism and redox metabolism, along with DNA repair mechanisms, were activated. The induction of hypoxia-regulated genes and the generalized regulation of multiple signaling pathways support the hypothesis that WAF exposition causes a hypoxia-like condition.
Collapse
Affiliation(s)
- Jesús Alejandro Zamora-Briseño
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Monica Améndola-Pimenta
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | | | - Alejandro Pereira-Santana
- División de Biotecnología Industrial, CONACYT-Centro de Investigación y Asistencia en Tecnología y Diseño del estado de Jalisco, Camino Arenero 1227, El Bajío, C.P. 45019, Zapopan, Jalisco, Mexico
| | - Ioreni Margarita Hernández-Velázquez
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Carlos Eduardo González-Penagos
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Juan Antonio Pérez-Vega
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Marcela Del Río-García
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Flor Árcega-Cabrera
- Unidad de Química Sisal, Facultad de Química, Universidad Nacional Autónoma de México, Puerto de Abrigo S/N, 97356, Sisal, Yucatán, Mexico
| | - Rossanna Rodríguez-Canul
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico.
- Laboratorio de Inmunología y Biología Molecular, CINVESTAV-IPN Unidad Mérida, Antigua carretera a Progreso Km 6., CP 97310, Mérida, Yucatán, Mexico.
| |
Collapse
|
65
|
Fattahi F, Saeednejad Zanjani L, Habibi Shams Z, Kiani J, Mehrazma M, Najafi M, Madjd Z. High expression of DNA damage-inducible transcript 4 (DDIT4) is associated with advanced pathological features in the patients with colorectal cancer. Sci Rep 2021; 11:13626. [PMID: 34211002 PMCID: PMC8249407 DOI: 10.1038/s41598-021-92720-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023] Open
Abstract
DNA damage-inducible transcript 4 (DDIT4) is induced in various cellular stress conditions. This study was conducted to investigate expression and prognostic significance of DDIT4 protein as a biomarker in the patients with colorectal cancer (CRC). PPI network and KEGG pathway analysis were applied to identify hub genes among obtained differentially expressed genes in CRC tissues from three GEO Series. In clinical, expression of DDIT4 as one of hub genes in three subcellular locations was evaluated in 198 CRC tissues using immunohistochemistry method on tissue microarrays. The association between DDIT4 expression and clinicopathological features as well as survival outcomes were analyzed. Results of bioinformatics analysis indicated 14 hub genes enriched in significant pathways according to KEGG pathways analysis among which DDIT4 was selected to evaluate CRC tissues. Overexpression of nuclear DDIT4 protein was found in CRC tissues compared to adjacent normal tissues (P = 0.003). Furthermore, higher nuclear expression of DDIT4 was found to be significantly associated with the reduced tumor differentiation and advanced TNM stages (all, P = 0.009). No significant association was observed between survival outcomes and nuclear expression of DDIT4 in CRC cases. Our findings indicated higher nuclear expression of DDIT4 was significantly associated with more aggressive tumor behavior and more advanced stage of disease in the patients with CRC.
Collapse
Affiliation(s)
- Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Mehrazma
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
66
|
Zhang G, Liu M, Chen H, Wu Z, Gao Y, Ma Z, He X, Kang X. NF-κB signalling pathways in nucleus pulposus cell function and intervertebral disc degeneration. Cell Prolif 2021; 54:e13057. [PMID: 34028920 PMCID: PMC8249791 DOI: 10.1111/cpr.13057] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/25/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is a common clinical degenerative disease of the spine. A series of factors, such as inflammation, oxidative stress and mechanical stress, promote degradation of the extracellular matrix (ECM) of the intervertebral discs (IVD), leading to dysfunction and structural destruction of the IVD. Nuclear factor-κB (NF-κB) transcription factor has long been regarded as a pathogenic factor of IDD. Therefore, NF-κB may be an ideal therapeutic target for IDD. As NF-κB is a multifunctional functional transcription factor with roles in a variety of biological processes, a comprehensive understanding of the function and regulatory mechanism of NF-κB in IDD pathology will be useful for the development of targeted therapeutic strategies for IDD, which can prevent the progression of IDD and reduce potential risks. This review discusses the role of the NF-κB signalling pathway in the nucleus pulposus (NP) in the process of IDD to understand pathological NP degeneration further and provide potential therapeutic targets that may interfere with NF-κB signalling for IDD therapy.
Collapse
Affiliation(s)
- Guang‐Zhi Zhang
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Ming‐Qiang Liu
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Hai‐Wei Chen
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Zuo‐Long Wu
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Yi‐Cheng Gao
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Zhan‐Jun Ma
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Xue‐Gang He
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
| | - Xue‐Wen Kang
- Department of OrthopedicsLanzhou University Second HospitalLanzhouChina
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Key Laboratory of Orthopedics Disease of Gansu ProvinceLanzhou University Second HospitalLanzhouChina
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal DisordersLanzhouChina
| |
Collapse
|
67
|
Niu M, Li L, Su Z, Wei L, Pu W, Zhao C, Ding Y, Wazir J, Cao W, Song S, Gao Q, Wang H. An integrative transcriptome study reveals Ddit4/Redd1 as a key regulator of cancer cachexia in rodent models. Cell Death Dis 2021; 12:652. [PMID: 34175899 PMCID: PMC8236061 DOI: 10.1038/s41419-021-03932-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
Cancer cachexia is a multifactorial metabolic syndrome that causes up to 20% of cancer-related deaths. Muscle atrophy, the hallmark of cancer cachexia, strongly impairs the quality of life of cancer patients; however, the underlying pathological process is still poorly understood. Investigation of the disease pathogenesis largely relies on cachectic mouse models. In our study, the transcriptome of the cachectic gastrocnemius muscle in the C26 xenograft model was integrated and compared with that of 5 more different datasets. The bioinformatic analysis revealed pivotal gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the disease, and the key genes were validated. Construction of the protein-protein interaction network and the comparison of pathways enriched in cancer cachexia with 5 other muscle atrophy models revealed Ddit4 (DNA damage-inducible transcript 4), as a key protein in cancer cachexia. The higher expression of Ddit4 in cachectic muscle was further validated in animal models and cachectic cancer patients. Further study revealed that p38 induced the expression of Ddit4, which in turn inhibited the mTOR pathway in atrophic cells.
Collapse
Affiliation(s)
- Mengyuan Niu
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Yibing Ding
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Wangsen Cao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China.
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China.
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School of Nanjing University, Nanjing, P. R. China.
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P. R. China.
| |
Collapse
|
68
|
Pérez-Sisqués L, Sancho-Balsells A, Solana-Balaguer J, Campoy-Campos G, Vives-Isern M, Soler-Palazón F, Anglada-Huguet M, López-Toledano MÁ, Mandelkow EM, Alberch J, Giralt A, Malagelada C. RTP801/REDD1 contributes to neuroinflammation severity and memory impairments in Alzheimer's disease. Cell Death Dis 2021; 12:616. [PMID: 34131105 PMCID: PMC8206344 DOI: 10.1038/s41419-021-03899-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death. Its downregulation in Parkinson's and Huntington's disease models ameliorates the pathological phenotypes. In the context of Alzheimer's disease (AD), the coding gene for RTP801, DDIT4, is responsive to Aβ and modulates its cytotoxicity in vitro. Also, RTP801 mRNA levels are increased in AD patients' lymphocytes. However, the involvement of RTP801 in the pathophysiology of AD has not been yet tested. Here, we demonstrate that RTP801 levels are increased in postmortem hippocampal samples from AD patients. Interestingly, RTP801 protein levels correlated with both Braak and Thal stages of the disease and with GFAP expression. RTP801 levels are also upregulated in hippocampal synaptosomal fractions obtained from murine 5xFAD and rTg4510 mice models of the disease. A local RTP801 knockdown in the 5xFAD hippocampal neurons with shRNA-containing AAV particles ameliorates cognitive deficits in 7-month-old animals. Upon RTP801 silencing in the 5xFAD mice, no major changes were detected in hippocampal synaptic markers or spine density. Importantly, we found an unanticipated recovery of several gliosis hallmarks and inflammasome key proteins upon neuronal RTP801 downregulation in the 5xFAD mice. Altogether our results suggest that RTP801 could be a potential future target for theranostic studies since it could be a biomarker of neuroinflammation and neurotoxicity severity of the disease and, at the same time, a promising therapeutic target in the treatment of AD.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Júlia Solana-Balaguer
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Genís Campoy-Campos
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marcel Vives-Isern
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ferran Soler-Palazón
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marta Anglada-Huguet
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | | | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain.
| | - Cristina Malagelada
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
69
|
Fattahi F, Kiani J, Alemrajabi M, Soroush A, Naseri M, Najafi M, Madjd Z. Overexpression of DDIT4 and TPTEP1 are associated with metastasis and advanced stages in colorectal cancer patients: a study utilizing bioinformatics prediction and experimental validation. Cancer Cell Int 2021; 21:303. [PMID: 34107956 PMCID: PMC8191213 DOI: 10.1186/s12935-021-02002-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Various diagnostic and prognostic tools exist in colorectal cancer (CRC) due to multiple genetic and epigenetic alterations causing the disease. Today, the expression of RNAs is being used as prognostic markers for cancer. METHODS In the current study, various dysregulated RNAs in CRC were identified via bioinformatics prediction. Expression of several of these RNAs were measured by RT-qPCR in 48 tissues from CRC patients as well as in colorectal cancer stem cell-enriched spheroids derived from the HT-29 cell line. The relationships between the expression levels of these RNAs and clinicopathological features were analyzed. RESULTS Our bioinformatics analysis determined 11 key mRNAs, 9 hub miRNAs, and 18 lncRNAs which among them 2 coding RNA genes including DDIT4 and SULF1 as well as 3 non-coding RNA genes including TPTEP1, miR-181d-5p, and miR-148b-3p were selected for the further investigations. Expression of DDIT4, TPTEP1, and miR-181d-5p showed significantly increased levels while SULF1 and miR-148b-3p showed decreased levels in CRC tissues compared to the adjacent normal tissues. Positive relationships between DDIT4, SULF1, and TPTEP1 expression and metastasis and advanced stages of CRC were observed. Additionally, our results showed significant correlations between expression of TPTEP1 with DDIT4 and SULF1. CONCLUSIONS Our findings demonstrated increased expression levels of DDIT4 and TPTEP1 in CRC were associated with more aggressive tumor behavior and more advanced stages of the disease. The positive correlations between TPTEP1 as non-coding RNA and both DDIT4 and SULF1 suggest a regulatory effect of TPTEP1 on these genes.
Collapse
Affiliation(s)
- Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Alemrajabi
- Firoozgar Clinical Research Development Center (FCRDC), Iran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Naseri
- Oncopathology Research Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
70
|
White JP. Amino Acid Trafficking and Skeletal Muscle Protein Synthesis: A Case of Supply and Demand. Front Cell Dev Biol 2021; 9:656604. [PMID: 34136478 PMCID: PMC8201612 DOI: 10.3389/fcell.2021.656604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/28/2021] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle protein synthesis is a highly complex process, influenced by nutritional status, mechanical stimuli, repair programs, hormones, and growth factors. The molecular aspects of protein synthesis are centered around the mTORC1 complex. However, the intricacies of mTORC1 regulation, both up and downstream, have expanded overtime. Moreover, the plastic nature of skeletal muscle makes it a unique tissue, having to coordinate between temporal changes in myofiber metabolism and hypertrophy/atrophy stimuli within a tissue with considerable protein content. Skeletal muscle manages the push and pull between anabolic and catabolic pathways through key regulatory proteins to promote energy production in times of nutrient deprivation or activate anabolic pathways in times of nutrient availability and anabolic stimuli. Branched-chain amino acids (BCAAs) can be used for both energy production and signaling to induce protein synthesis. The metabolism of BCAAs occur in tandem with energetic and anabolic processes, converging at several points along their respective pathways. The fate of intramuscular BCAAs adds another layer of regulation, which has consequences to promote or inhibit muscle fiber protein anabolism. This review will outline the general mechanisms of muscle protein synthesis and describe how metabolic pathways can regulate this process. Lastly, we will discuss how BCAA availability and demand coordinate with synthesis mechanisms and identify key factors involved in intramuscular BCAA trafficking.
Collapse
Affiliation(s)
- James P White
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States.,Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States.,Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
71
|
Örd T, Örd D, Kaikkonen MU, Örd T. Pharmacological or TRIB3-Mediated Suppression of ATF4 Transcriptional Activity Promotes Hepatoma Cell Resistance to Proteasome Inhibitor Bortezomib. Cancers (Basel) 2021; 13:cancers13102341. [PMID: 34066165 PMCID: PMC8150958 DOI: 10.3390/cancers13102341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/17/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Proteasome inhibitors are currently used in the treatment of certain blood cancers, and clinical trials to treat solid tumors, including liver cancer, have also been conducted. However, different malignancies are not equally susceptible to proteasome inhibitors, and resistance to the drug may develop during the therapy. Here, we characterize the molecular mechanisms underlying the resilience of liver cancer cells to the proteasome inhibitor bortezomib. The results demonstrate that the activity of the eIF2α–ATF4 stress response pathway affects the viability of cells treated with bortezomib. We found that the pseudokinase TRIB3, an endogenous regulator of ATF4 and a gene highly expressed in liver cancer, resides predominantly at the same chromatin sites as ATF4 and constrains ATF4 activity. The survival of bortezomib-exposed hepatoma cells proved sensitive to TRIB3 overexpression and inactivation. Thus, TRIB3 is a novel factor contributing to bortezomib resistance of liver cancer cells. Abstract The proteasome is an appealing target for anticancer therapy and the proteasome inhibitor bortezomib has been approved for the treatment of several types of malignancies. However, the molecular mechanisms underlying cancer cell resistance to bortezomib remain poorly understood. In the current article, we investigate how modulation of the eIF2α–ATF4 stress pathway affects hepatoma cell response to bortezomib. Transcriptome profiling revealed that many ATF4 transcriptional target genes are among the most upregulated genes in bortezomib-treated HepG2 human hepatoma cells. While pharmacological enhancement of the eIF2α–ATF4 pathway activity results in the elevation of the activities of all branches of the unfolded protein response (UPR) and sensitizes cells to bortezomib toxicity, the suppression of ATF4 induction delays bortezomib-induced cell death. The pseudokinase TRIB3, an inhibitor of ATF4, is expressed at a high basal level in hepatoma cells and is strongly upregulated in response to bortezomib. To map genome-wide chromatin binding loci of TRIB3 protein, we fused a Flag tag to endogenous TRIB3 in HepG2 cells and performed ChIP-Seq. The results demonstrate that TRIB3 predominantly colocalizes with ATF4 on chromatin and binds to genomic regions containing the C/EBP–ATF motif. Bortezomib treatment leads to a robust enrichment of TRIB3 binding near genes induced by bortezomib and involved in the ER stress response and cell death. Disruption of TRIB3 increases C/EBP–ATF-driven transcription, augments ER stress and cell death upon exposure to bortezomib, while TRIB3 overexpression enhances cell survival. Thus, TRIB3, colocalizing with ATF4 and limiting its transcriptional activity, functions as a factor increasing resistance to bortezomib, while pharmacological over-activation of eIF2α–ATF4 can overcome the endogenous restraint mechanisms and sensitize cells to bortezomib.
Collapse
Affiliation(s)
- Tiit Örd
- Institute of Genomics, University of Tartu, Riia 23b, 51010 Tartu, Estonia; (T.Ö.); (D.Ö.)
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland;
| | - Daima Örd
- Institute of Genomics, University of Tartu, Riia 23b, 51010 Tartu, Estonia; (T.Ö.); (D.Ö.)
| | - Minna U. Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland;
| | - Tõnis Örd
- Institute of Genomics, University of Tartu, Riia 23b, 51010 Tartu, Estonia; (T.Ö.); (D.Ö.)
- Correspondence:
| |
Collapse
|
72
|
Pérez-Sisqués L, Martín-Flores N, Masana M, Solana-Balaguer J, Llobet A, Romaní-Aumedes J, Canal M, Campoy-Campos G, García-García E, Sánchez-Fernández N, Fernández-García S, Gilbert JP, Rodríguez MJ, Man HY, Feinstein E, Williamson DL, Soto D, Gasull X, Alberch J, Malagelada C. RTP801 regulates motor cortex synaptic transmission and learning. Exp Neurol 2021; 342:113755. [PMID: 33984337 DOI: 10.1016/j.expneurol.2021.113755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/24/2021] [Accepted: 05/08/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death in in vitro and in vivo models of Parkinson's and Huntington's diseases and is up regulated in compromised neurons in human postmortem brains of both neurodegenerative disorders. Indeed, in both Parkinson's and Huntington's disease mouse models, RTP801 knockdown alleviates motor-learning deficits. RESULTS We investigated the physiological role of RTP801 in neuronal plasticity and we found RTP801 in rat, mouse and human synapses. The absence of RTP801 enhanced excitatory synaptic transmission in both neuronal cultures and brain slices from RTP801 knock-out (KO) mice. Indeed, RTP801 KO mice showed improved motor learning, which correlated with lower spine density but increased basal filopodia and mushroom spines in the motor cortex layer V. This paralleled with higher levels of synaptosomal GluA1 and TrkB receptors in homogenates derived from KO mice motor cortex, proteins that are associated with synaptic strengthening. CONCLUSIONS Altogether, these results indicate that RTP801 has an important role modulating neuronal plasticity and motor learning. They will help to understand its role in neurodegenerative disorders where RTP801 levels are detrimentally upregulated.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain.
| | - Núria Martín-Flores
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Mercè Masana
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Júlia Solana-Balaguer
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Arnau Llobet
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain
| | - Joan Romaní-Aumedes
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Mercè Canal
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Genís Campoy-Campos
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Esther García-García
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | | | - Sara Fernández-García
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - James P Gilbert
- Department of Biology, Pharmacology and Experimental Therapeutics, Boston University, Boston, MA 02215, USA
| | - Manuel José Rodríguez
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Heng-Ye Man
- Department of Biology, Pharmacology and Experimental Therapeutics, Boston University, Boston, MA 02215, USA
| | | | - David L Williamson
- Kinesiology Program, School of Behavioral Sciences and Education, Penn State Harrisburg, Middletown, PA 17057, USA
| | - David Soto
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain
| | - Xavier Gasull
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain
| | - Jordi Alberch
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Cristina Malagelada
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain.
| |
Collapse
|
73
|
DDIT4 Novel Mutations in Pancreatic Cancer. Gastroenterol Res Pract 2021; 2021:6674404. [PMID: 34007269 PMCID: PMC8110378 DOI: 10.1155/2021/6674404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 11/18/2022] Open
Abstract
Pancreatic cancer is one of the most common malignancies worldwide. This study is aimed at searching the possible genetic mutations and the value of novel gene mutation in the DNA damage-inducible transcript 4 (DDIT4) and signaling pathway in pancreatic cancer. Polymerase chain reaction (PCR) was performed to amplify the DNA sequences of DDIT4 from patients with pancreatic ductal adenocarcinoma. In addition, we used IHC to detect the expression level of DDIT4 in patients with pancreatic cancer in different types of gene mutation. Double-labeled immunofluorescence was employed to explore the expression levels of DDIT4/LC3 and their potential correlation. Our work indicated the two novel stable gene mutations in DDIT4 mRNA 3'-untranslated region (m.990 U>A and m.1246 C>U). Thirteen samples were found to have mutation in the DDIT4 3'-untranslated regions (UTR). To further verify the influence of gene mutation on protein expression, we performed immunohistochemistry on different gene mutation types, and we found a correlation between DDIT4 expression and gene mutation, which is accompanied by nuclear staining deepening. In order to further discuss the clinical value of DDIT4 gene mutation, immunofluorescence suggested that the expression of DDIT4 colocated with LC3; thus, we speculated that DDIT4 mutation may be involved in autophagy in pancreatic cancer cell. In this study, we found mutation in the 3'-UTR region of DDIT4, which may be associated with DDIT4 expression and tumor autophagy in pancreatic cancer tissues.
Collapse
|
74
|
Exploiting the Role of Hypoxia-Inducible Factor 1 and Pseudohypoxia in the Myelodysplastic Syndrome Pathophysiology. Int J Mol Sci 2021; 22:ijms22084099. [PMID: 33921064 PMCID: PMC8071466 DOI: 10.3390/ijms22084099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal hematopoietic stem (HSCs) and/or progenitor cells disorders. The established dependence of MDS progenitors on the hypoxic bone marrow (BM) microenvironment turned scientific interests to the transcription factor hypoxia-inducible factor 1 (HIF-1). HIF-1 facilitates quiescence maintenance and regulates differentiation by manipulating HSCs metabolism, being thus an appealing research target. Therefore, we examine the aberrant HIF-1 stabilization in BMs from MDS patients and controls (CTRLs). Using a nitroimidazole–indocyanine conjugate, we show that HIF-1 aberrant expression and transcription activity is oxygen independent, establishing the phenomenon of pseudohypoxia in MDS BM. Next, we examine mitochondrial quality and quantity along with levels of autophagy in the differentiating myeloid lineage isolated from fresh BM MDS and CTRL aspirates given that both phenomena are HIF-1 dependent. We show that the mitophagy of abnormal mitochondria and autophagic death are prominently featured in the MDS myeloid lineage, their severity increasing with intra-BM blast counts. Finally, we use in vitro cultured CD34+ HSCs isolated from fresh human BM aspirates to manipulate HIF-1 expression and examine its potential as a therapeutic target. We find that despite being cultured under 21% FiO2, HIF-1 remained aberrantly stable in all MDS cultures. Inhibition of the HIF-1α subunit had a variable beneficial effect in all <5%-intra-BM blasts-MDS, while it had no effect in CTRLs or in ≥5%-intra-BM blasts-MDS that uniformly died within 3 days of culture. We conclude that HIF-1 and pseudohypoxia are prominently featured in MDS pathobiology, and their manipulation has some potential in the therapeutics of benign MDS.
Collapse
|
75
|
Andrysik Z, Bender H, Galbraith MD, Espinosa JM. Multi-omics analysis reveals contextual tumor suppressive and oncogenic gene modules within the acute hypoxic response. Nat Commun 2021; 12:1375. [PMID: 33654095 PMCID: PMC7925689 DOI: 10.1038/s41467-021-21687-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular adaptation to hypoxia is a hallmark of cancer, but the relative contribution of hypoxia-inducible factors (HIFs) versus other oxygen sensors to tumorigenesis is unclear. We employ a multi-omics pipeline including measurements of nascent RNA to characterize transcriptional changes upon acute hypoxia. We identify an immediate early transcriptional response that is strongly dependent on HIF1A and the kinase activity of its cofactor CDK8, includes indirect repression of MYC targets, and is highly conserved across cancer types. HIF1A drives this acute response via conserved high-occupancy enhancers. Genetic screen data indicates that, in normoxia, HIF1A displays strong cell-autonomous tumor suppressive effects through a gene module mediating mTOR inhibition. Conversely, in advanced malignancies, expression of a module of HIF1A targets involved in collagen remodeling is associated with poor prognosis across diverse cancer types. In this work, we provide a valuable resource for investigating context-dependent roles of HIF1A and its targets in cancer biology. The response to hypoxia can significantly impact oncogenic processes. Here, the authors define the early transcriptional response to acute hypoxia and identify HIF1A target genes as part of this acute response, providing a resource for investigating context-dependent roles of HIF1A in the biology of cancer.
Collapse
Affiliation(s)
- Zdenek Andrysik
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Heather Bender
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Galbraith
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. .,Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Joaquin M Espinosa
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. .,Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA. .,Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
76
|
Miller WP, Sunilkumar S, Dennis MD. The stress response protein REDD1 as a causal factor for oxidative stress in diabetic retinopathy. Free Radic Biol Med 2021; 165:127-136. [PMID: 33524531 PMCID: PMC7956244 DOI: 10.1016/j.freeradbiomed.2021.01.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Diabetic Retinopathy (DR) is a major cause of visual dysfunction, yet much remains unknown regarding the specific molecular events that contribute to diabetes-induced retinal pathophysiology. Herein, we review the impact of oxidative stress on DR, and explore evidence that supports a key role for the stress response protein regulated in development and DNA damage (REDD1) in the development of diabetes-induced oxidative stress and functional defects in vision. It is well established that REDD1 mediates the cellular response to a number of diverse stressors through repression of the central metabolic regulator known as mechanistic target of rapamycin complex 1 (mTORC1). A growing body of evidence also supports that REDD1 acts independent of mTORC1 to promote oxidative stress by both enhancing the production of reactive oxygen species and suppressing the antioxidant response. Collectively, there is strong preclinical data to support a key role for REDD1 in the development and progression of retinal complications caused by diabetes. Furthermore, early proof-of-concept clinical trials have found a degree of success in combating ischemic retinal disease through intravitreal delivery of an siRNA targeting the REDD1 mRNA. Overall, REDD1-associated signaling represents an intriguing target for novel clinical therapies that go beyond addressing the symptoms of diabetes by targeting the underlying molecular mechanisms that contribute to DR.
Collapse
Affiliation(s)
- William P Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Siddharth Sunilkumar
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA; Department of Ophthalmology, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
77
|
Mundo W, Wolfson G, Moore LG, Houck JA, Park D, Julian CG. Hypoxia-induced inhibition of mTORC1 activity in the developing lung: a possible mechanism for the developmental programming of pulmonary hypertension. Am J Physiol Heart Circ Physiol 2021; 320:H980-H990. [PMID: 33416457 PMCID: PMC7988757 DOI: 10.1152/ajpheart.00520.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/14/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Perinatal hypoxia induces permanent structural and functional changes in the lung and its pulmonary circulation that are associated with the development of pulmonary hypertension (PH) in later life. The mechanistic target of the rapamycin (mTOR) pathway is vital for fetal lung development and is implicated in hypoxia-associated PH, yet its involvement in the developmental programming of PH remains unclear. Pregnant C57/BL6 dams were placed in hyperbaric (760 mmHg) or hypobaric chambers during gestation (505 mmHg, day 15 through postnatal day 4) or from weaning through adulthood (420 mmHg, postnatal day 21 through 8 wk). Pulmonary hemodynamics and right ventricular systolic pressure (RVSP) were measured at 8 wk. mTOR pathway proteins were assessed in fetal (day 18.5) and adult lung (8 wk). Perinatal hypoxia induced PH during adulthood, even in the absence of a sustained secondary hypoxic exposure, as indicated by reduced pulmonary artery acceleration time (PAAT) and peak flow velocity through the pulmonary valve, as well as greater RVSP, right ventricular (RV) wall thickness, and RV/left ventricular (LV) weight. Such effects were independent of increased blood viscosity. In fetal lung homogenates, hypoxia reduced the expression of critical downstream mTOR targets, most prominently total and phosphorylated translation repressor protein (4EBP1), as well as vascular endothelial growth factor, a central regulator of angiogenesis in the fetal lung. In contrast, adult offspring of hypoxic dams tended to have elevated p4EBP1 compared with controls. Our data suggest that inhibition of mTORC1 activity in the fetal lung as a result of gestational hypoxia may interrupt pulmonary vascular development and thereby contribute to the developmental programming of PH.NEW & NOTEWORTHY We describe the first study to evaluate a role for the mTOR pathway in the developmental programming of pulmonary hypertension. Our findings suggest that gestational hypoxia impairs mTORC1 activation in the fetal lung and may impede pulmonary vascular development, setting the stage for pulmonary vascular disease in later life.
Collapse
Affiliation(s)
- William Mundo
- School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Gabriel Wolfson
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Do Park
- School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
78
|
Savukaitytė A, Gudoitytė G, Bartnykaitė A, Ugenskienė R, Juozaitytė E. siRNA Knockdown of REDD1 Facilitates Aspirin-Mediated Dephosphorylation of mTORC1 Target 4E-BP1 in MDA-MB-468 Human Breast Cancer Cell Line. Cancer Manag Res 2021; 13:1123-1133. [PMID: 33574709 PMCID: PMC7872862 DOI: 10.2147/cmar.s264414] [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: 06/17/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mutations within genes encoding components of the PI3K/AKT/mTOR (phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin) signaling axis frequently activate the pathway in breast cancer, making it an attractive therapeutic target. Inhibition of mTORC1 (mechanistic target of rapamycin complex 1) activity upon aspirin treatment has been reported in breast cancer cells harboring PI3KCA (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha) mutation and is considered to account for anticancer action. Methods MDA-MB-468 (harbors mutated PTEN (phosphatase and TENsin homolog)), MCF-7 (PI3KCA-mutated), MDA-MB-231 (no PI3K pathway mutations) cancer cell lines and MCF10A non-cancerous breast epithelial cells were employed for the assessment of modulation of mTORC1 signaling by aspirin. Targeted amplicon-based next-generation sequencing using the Ion Torrent technology was carried out to determine gene expression changes following drug treatment. Western blot was performed to analyze the expression and phosphorylation of proteins. Knockdown by siRNA approach was applied to assess the role of REDD1/DDIT4 (DNA damage-inducible transcript 4) in mTORC1 inhibition by aspirin. Results We show a decline in phosphorylation of mTORC1 downstream substrate 4E-BP1 (eukaryotic translation initiation factor 4E-binding protein 1) in response to treatment with aspirin and its metabolite salicylic acid in MDA-MB-468, MCF-7, MDA-MB-231, and MCF10A cell lines. We further demonstrate a novel molecular response to aspirin in breast cancer cells. Specifically, we found that aspirin and salicylic acid increase the expression of REDD1 protein, that is known for its suppressive function towards mTORC1. Unexpectedly, we observed that siRNA knockdown of REDD1 expression facilitated aspirin-mediated suppression of mTORC1 downstream substrate 4E-BP1 phosphorylation in the MDA-MB-468 cell line. REDD1 downregulation slightly encouraged reduction in 4E-BP1 phosphorylation by aspirin in MCF-7 cells but did not elicit a reproducible effect in the MDA-MB-231 cell line. siRNA knockdown of REDD1 did not affect the expression of phosphorylated form of 4E-BP1 following aspirin treatment in MCF10A non-cancerous breast epithelial cells. Conclusion The current findings suggest that REDD1 downregulation might improve the anticancer activity of aspirin in a subset of breast tumors.
Collapse
Affiliation(s)
- Aistė Savukaitytė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Greta Gudoitytė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Agnė Bartnykaitė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Ugenskienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Institute of Biology Systems and Genetic Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Elona Juozaitytė
- Department of Oncology and Hematology, Hospital of Lithuanian University of Health Sciences Kaunas Clinics, Kaunas, Lithuania
| |
Collapse
|
79
|
Lu G, Wu Z, Shang J, Xie Z, Chen C, Zhang C. The effects of metformin on autophagy. Biomed Pharmacother 2021; 137:111286. [PMID: 33524789 DOI: 10.1016/j.biopha.2021.111286] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Metformin is the first-line option for treating newly diagnosed diabetic patients and also involved in other pharmacological actions, including antitumor effect, anti-aging effect, polycystic ovarian syndrome prevention, cardiovascular action, and neuroprotective effect, etc. However, the mechanisms of metformin actions were not fully illuminated. Recently, increasing researches showed that autophagy is a vital medium of metformin playing pharmacological actions. Nevertheless, results on the effects of metformin on autophagy were inconsistent. Apart from few clinical evidences, more data focused on kinds of no-clinical models. First, many studies showed that metformin could induce autophagy via a number of signaling pathways, including AMPK-related signaling pathways (e.g. AMPK/mTOR, AMPK/CEBPD, MiTF/TFE, AMPK/ULK1, and AMPK/miR-221), Redd1/mTOR, STAT, SIRT, Na+/H+ exchangers, MAPK/ERK, PK2/PKR/AKT/ GSK3β, and TRIB3. Secondly, some signaling pathways were involved in the process of metformin inhibiting autophagy, such as AMPK-related signaling pathways (AMPK/NF-κB and other undetermined AMPK-related signaling pathways), Hedgehog, miR-570-3p, miR-142-3p, and MiR-3127-5p. Thirdly, two types of signaling pathways including PI3K/AKT/mTOR and endoplasmic reticulum (ER) stress could bidirectionally impact the effectiveness of metformin on autophagy. Finally, multiple signal pathways were reviewed collectively in terms of affecting the effectiveness of metformin on autophagy. The pharmacological effects of metformin combining its actions on autophagy were also discussed. It would help better apply metformin to treat diseases in term of mediating autophagy.
Collapse
Affiliation(s)
- Guangli Lu
- School of Business, Henan University, Henan, Kaifeng, China
| | - Zhen Wu
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China
| | - Jia Shang
- School of Kaifeng Culture and Tourism, Henan, Kaifeng, China
| | - Zhenxing Xie
- School of Basic Medicine, Henan University, Henan, Kaifeng, Jinming Avenue, 475004, China.
| | - Chaoran Chen
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China.
| | - Chuning Zhang
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Henan, Kaifeng, China
| |
Collapse
|
80
|
Korkmaz C, Cansu DU, Cansu GB. Familial Mediterranean fever: the molecular pathways from stress exposure to attacks. Rheumatology (Oxford) 2021; 59:3611-3621. [PMID: 33026080 DOI: 10.1093/rheumatology/keaa450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/21/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
FMF is an autoinflammatory disease characterized by recurrent attacks and increased IL-1 synthesis owing to activation of the pyrin inflammasome. Although knowledge of the mechanisms leading to the activation of pyrin inflammasome is increasing, it is still unknown why the disease is characterized by attack. The emergence of FMF attacks after emotional stress and the induction of attacks with metaraminol in previous decades suggested that stress-induced sympathoadrenal system activation might play a role in inflammasome activation and triggering attacks. In this review, we will review the possible molecular mechanism of stress mediators on the inflammation pathway and inflammasome activation. Studies on stress mediators and their impact on inflammation pathways will provide a better understanding of stress-related exacerbation mechanisms in both autoinflammatory and autoimmune diseases. This review provides a new perspective on this subject and will contribute to new studies.
Collapse
Affiliation(s)
- Cengiz Korkmaz
- Division of Rheumatology, Department of Internal Medicine, Eskisehir Osmangazi University, School of Medicine, Eskisehir
| | - Döndü U Cansu
- Division of Rheumatology, Department of Internal Medicine, Eskisehir Osmangazi University, School of Medicine, Eskisehir
| | - Güven Barış Cansu
- Department of Endocrinology, Kütahya Health Science University, School of Medicine, Kütahya, 43100, Turkey
| |
Collapse
|
81
|
Discovery of a new molecule inducing melanoma cell death: dual AMPK/MELK targeting for novel melanoma therapies. Cell Death Dis 2021; 12:64. [PMID: 33431809 PMCID: PMC7801734 DOI: 10.1038/s41419-020-03344-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
In the search of biguanide-derived molecules against melanoma, we have discovered and developed a series of bioactive products and identified the promising new compound CRO15. This molecule exerted anti-melanoma effects on cells lines and cells isolated from patients including the ones derived from tumors resistant to BRAF inhibitors. Moreover, CRO15 was able to decrease viability of cells lines from a broad range of cancer types. This compound acts by two distinct mechanisms. First by activating the AMPK pathway induced by a mitochondrial disorder. Second by inhibition of MELK kinase activity, which induces cell cycle arrest and activation of DNA damage repair pathways by p53 and REDD1 activation. All of these mechanisms activate autophagic and apoptotic processes resulting in melanoma cell death. The strong efficacy of CRO15 to reduce the growth of melanoma xenograft sensitive or resistant to BRAF inhibitors opens interesting perspective.
Collapse
|
82
|
Ding F, Gao F, Zhang S, Lv X, Chen Y, Liu Q. A review of the mechanism of DDIT4 serve as a mitochondrial related protein in tumor regulation. Sci Prog 2021; 104:36850421997273. [PMID: 33729069 PMCID: PMC10455034 DOI: 10.1177/0036850421997273] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DDIT4 is a mitochondrial and tumor-related protein involved in anti-tumor therapy resistance, proliferation, and invasion, etc. Its expression level increases under the stress such as chemotherapy, hypoxia, and DNA damage. A number of clinical studies have confirmed that DDIT4 can change the behavior of tumor cells and the prognosis of patients with cancer. However, the role of DDIT4 in promoting or suppressing cancer is still inconclusive. This article summarized the four characteristics of DDIT4 including a mitochondria-related protein, interactions with various protein molecules, immune and metabolic cell related proteins and participator in the oxygen sensing pathway, which may be related to the progress of cancer.
Collapse
Affiliation(s)
- Fadian Ding
- Department of Hepatobiliary Pancreatic Surgery, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Feng Gao
- Department of Pathology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Sheng Zhang
- Department of Pathology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoting Lv
- Department of Respiratory, First Affiliated Hospital; Fujian Medical University, Fuzhou, China
| | - Youting Chen
- Department of Hepatobiliary Pancreatic Surgery, 1st Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qicai Liu
- Center for Reproductive Medicine, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
83
|
Clapper E, Di Trapani G, Tonissen KF. The regulation of bcr-abl in hypoxia is through the mTOR pathway. Leuk Lymphoma 2020; 62:967-978. [PMID: 33251904 DOI: 10.1080/10428194.2020.1849679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is usually characterized by the formation of the fusion onco-protein bcr-abl. Therefore, the majority of CML treatments are bcr-abl specific tyrosine kinase inhibitors (TKIs). TKI resistance in CML treatment is becoming a major obstacle in managing this disease. One well-studied form of drug resistance is hypoxia-induced drug resistance, a phenomenon observed in many other cancers. This study aimed to determine the efficacy of TKIs in CML cells cultured in hypoxia. It was observed that bcr-abl translation was severely halted in hypoxia, rendering TKIs ineffective. We found that the mechanism by which bcr-abl protein levels were being suppressed in hypoxia was through the mTOR pathway, specifically via ribosomal protein S6 (RPS6). This information is vital to the improvement of CML treatments, as it can be used to determine how to best combat hypoxia-induced drug resistance in CML and subsequently to identify new targets for treatment.
Collapse
Affiliation(s)
- Erin Clapper
- School of Environment and Science, Griffith University, Nathan, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | | | - Kathryn F Tonissen
- School of Environment and Science, Griffith University, Nathan, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| |
Collapse
|
84
|
Embryonic exposure to hyper glucocorticoids suppresses brown fat development and thermogenesis via REDD1. Sci Bull (Beijing) 2020; 66:478-489. [PMID: 33936858 DOI: 10.1016/j.scib.2020.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Maternal stress during pregnancy is prevailing worldwide, which exposes fetuses to intrauterine hyper glucocorticoids (GC), programming offspring to obesity and metabolic diseases. Despite the importance of brown adipose tissue (BAT) in maintaining long-term metabolic health, impacts of prenatal hyper GC on postnatal BAT thermogenesis and underlying regulations remain poorly defined. Pregnant mice were administrated with synthetic GC dexamethasone (DEX) at levels comparable to fetal GC exposure of stressed mothers. Prenatal GC exposure dose-dependently reduced BAT thermogenic activity, contributing to lower body temperature and higher mortality of neonates; such difference was abolished under thermoneutrality, underscoring BAT deficiency was the major contributor to adverse changes in postnatal thermogenesis due to excessive GC. Prenatal GC exposure highly activated Redd1 expression and reduced Ppargc1a transcription from the alternative promoter (Ppargc1a-AP) in neonatal BAT. During brown adipocyte differentiation, ectopic Redd1 expression reduced Ppargc1a-AP expression and mitochondrial biogenesis; and the inhibitory effects of GC on mitochondrial biogenesis and Ppargc1a-AP expression were blocked by Redd1 ablation. Redd1 reduced protein kinase A phosphorylation and suppressed cyclic adenosine monophosphate (cAMP) -responsive element-binding protein (CREB) binding to the cAMP regulatory element (CRE) in Ppargc1a-AP promoter, leading to Ppargc1a-AP inactivation. In summary, excessive maternal GC exposure during pregnancy dysregulates Redd1-Ppargc1a-AP axis, which impairs fetal BAT development, hampering postnatal thermogenic adaptation and metabolic health of offspring.
Collapse
|
85
|
Issara-Amphorn J, Chancharoenthana W, Visitchanakun P, Leelahavanichkul A. Syk Inhibitor Attenuates Polymicrobial Sepsis in FcgRIIb-Deficient Lupus Mouse Model, the Impact of Lupus Characteristics in Sepsis. J Innate Immun 2020; 12:461-479. [PMID: 32927460 PMCID: PMC7747092 DOI: 10.1159/000509111] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/27/2020] [Indexed: 12/14/2022] Open
Abstract
The impact of spleen tyrosine kinase (Syk) signaling might be prominent in lupus because (i) Syk is a shared downstream signaling molecule among circulating immune complex, LPS, and (1→3)-β-D-glucan (BG), and (ii) all of these factors are detectable in the serum of Fc gamma receptor IIb-deficient (FcgRIIb-/-) mice with sepsis. As a proof of concept study, we activated macrophages with BG combined with LPS (BG + LPS). We found that BG + LPS predominantly upregulated Syk expression and proinflammatory cytokines in FcgRIIb-/- macrophages compared with wild-type (WT) macrophages. Syk inhibition downregulated several inflammatory pathways in FcgRIIb-/- macrophages activated with BG + LPS, as determined by RNA sequencing analysis, suggesting the potential anti-inflammatory impact of Syk inhibitors in lupus. Indeed, administration of a Syk inhibitor prior to cecal ligation and puncture (CLP) sepsis in FcgRIIb-/- mice reduced baseline lupus-induced proinflammatory cytokines and attenuated sepsis severity as evaluated by mortality, organ injury, serum LPS, and post-sepsis serum cytokines. In conclusion, it was easier to induce Syk expression in FcgRIIb-/- macrophages than in WT macrophages. This might be because of the loss of inhibitory signaling, which might be responsible for prominent Syk abundance in the spleens of 40-week-old FcgRIIb-/- mice and the potent effect of Syk inhibitor in lupus mice compared with WT.
Collapse
Affiliation(s)
- Jiraphorn Issara-Amphorn
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wiwat Chancharoenthana
- Nephrology Research Unit, Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Peerapat Visitchanakun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand,
| |
Collapse
|
86
|
Chen Y, Zhou X. Research progress of mTOR inhibitors. Eur J Med Chem 2020; 208:112820. [PMID: 32966896 DOI: 10.1016/j.ejmech.2020.112820] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/16/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a highly conserved Serine/Threonine (Ser/Thr) protein kinase, which belongs to phosphatidylinositol-3-kinase-related kinase (PIKK) protein family. mTOR exists as two types of protein complex: mTORC1 and mTORC2, which act as central controller regulating processes of cell metabolism, growth, proliferation, survival and autophagy. The mTOR inhibitors block mTOR signaling pathway, producing anti-inflammatory, anti-proliferative, autophagy and apoptosis induction effects, thus mTOR inhibitors are mainly used in cancer therapy. At present, mTOR inhibitors are divided into four categories: Antibiotic allosteric mTOR inhibitors (first generation), ATP-competitive mTOR inhibitors (second generation), mTOR/PI3K dual inhibitors (second generation) and other new mTOR inhibitors (third generation). In this article, these four categories of mTOR inhibitors and their structures, properties and some clinical researches will be introduced. Among them, we focus on the structure of mTOR inhibitors and try to analyze the structure-activity relationship. mTOR inhibitors are classified according to their chemical structure and their contents are introduced systematically. Moreover, some natural products that have direct or indirect mTOR inhibitory activities are introduced together. In this article, we analyzed the target, binding mode and structure-activity relationship of each generation of mTOR inhibitors and proposed two hypothetic scaffolds (the inverted-Y-shape scaffold and the C-shape scaffold) for the second generation of mTOR inhibitors. These findings may provide some help or reference for drug designing, drug modification or the future development of mTOR inhibitor.
Collapse
Affiliation(s)
- Yifan Chen
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xiaoping Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
87
|
Britto FA, Dumas K, Giorgetti-Peraldi S, Ollendorff V, Favier FB. Is REDD1 a metabolic double agent? Lessons from physiology and pathology. Am J Physiol Cell Physiol 2020; 319:C807-C824. [PMID: 32877205 DOI: 10.1152/ajpcell.00340.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Akt/mechanistic target of rapamycin (mTOR) signaling pathway governs macromolecule synthesis, cell growth, and metabolism in response to nutrients and growth factors. Regulated in development and DNA damage response (REDD)1 is a conserved and ubiquitous protein, which is transiently induced in response to multiple stimuli. Acting like an endogenous inhibitor of the Akt/mTOR signaling pathway, REDD1 protein has been shown to regulate cell growth, mitochondrial function, oxidative stress, and apoptosis. Recent studies also indicate that timely REDD1 expression limits Akt/mTOR-dependent synthesis processes to spare energy during metabolic stresses, avoiding energy collapse and detrimental consequences. In contrast to this beneficial role for metabolic adaptation, REDD1 chronic expression appears involved in the pathogenesis of several diseases. Indeed, REDD1 expression is found as an early biomarker in many pathologies including inflammatory diseases, cancer, neurodegenerative disorders, depression, diabetes, and obesity. Moreover, prolonged REDD1 expression is associated with cell apoptosis, excessive reactive oxygen species (ROS) production, and inflammation activation leading to tissue damage. In this review, we decipher several mechanisms that make REDD1 a likely metabolic double agent depending on its duration of expression in different physiological and pathological contexts. We also discuss the role played by REDD1 in the cross talk between the Akt/mTOR signaling pathway and the energetic metabolism.
Collapse
Affiliation(s)
| | - Karine Dumas
- Université Cote d'Azur, INSERM, UMR1065, C3M, Nice, France
| | | | | | | |
Collapse
|
88
|
Identification of a circRNA-miRNA-mRNA network to explore the effects of circRNAs on pathogenesis and treatment of spinal cord injury. Life Sci 2020; 257:118039. [DOI: 10.1016/j.lfs.2020.118039] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/13/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
|
89
|
Rivera-Gonzalez GC, Klopot A, Sabin K, Baida G, Horsley V, Budunova I. Regulated in Development and DNA Damage Responses 1 Prevents Dermal Adipocyte Differentiation and Is Required for Hair Cycle-Dependent Dermal Adipose Expansion. J Invest Dermatol 2020; 140:1698-1705.e1. [PMID: 32032578 PMCID: PMC7398827 DOI: 10.1016/j.jid.2019.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/14/2019] [Accepted: 12/04/2019] [Indexed: 02/07/2023]
Abstract
Dermal white adipose tissue (dWAT) expansion is associated with important homeostatic and pathologic processes in skin. Even though mTOR/protein kinase B signaling is important for adipogenesis, the role of regulated development of DNA damage responses 1 (REDD1), a negative regulator of mTOR/protein kinase B, is poorly understood. Loss of REDD1 in mice resulted in reduction of body mass, total fat, size of gonadal white adipose tissue, and interscapular brown adipose tissue. Inguinal subcutaneous white adipose tissue and dWAT in REDD1 knockouts were expanded compared with wild type mice. Size and number of mature adipocytes in dWAT were also increased in adult REDD1 knockouts. This dWAT phenotype was established around postnatal day 18 and did not depend on the hair growth cycle. Numbers of adipocyte precursor cells were lower in REDD1 knockout skin. In vitro analysis revealed increased differentiation of skin-derived REDD1 knockout adipocyte precursor cells as indicated by higher lipid accumulation and increased adipogenic marker expression. 3T3L1 cells overexpressing REDD1 had decreased sensitivity to differentiation. Overall, our findings indicate that REDD1 silencing induced expansion of dWAT through hypertrophy and hyperplasia. This REDD1-dependent mechanism of adipogenesis could be used to preferentially target skin-associated adipose tissue for therapeutic purposes.
Collapse
Affiliation(s)
- Guillermo C. Rivera-Gonzalez
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
- Current address: Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110
| | - Anna Klopot
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Kaitlyn Sabin
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
| | - Gleb Baida
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology and Department of Dermatology, Yale University, New Haven, CT 06520
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| |
Collapse
|
90
|
Kraus RJ, Cordes BLA, Sathiamoorthi S, Patel P, Yuan X, Iempridee T, Yu X, Lee DL, Lambert PF, Mertz JE. Reactivation of Epstein-Barr Virus by HIF-1α Requires p53. J Virol 2020; 94:e00722-20. [PMID: 32641480 PMCID: PMC7459560 DOI: 10.1128/jvi.00722-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
We previously reported that the cellular transcription factor hypoxia-inducible factor 1α (HIF-1α) binds a hypoxia response element (HRE) located within the promoter of Epstein-Barr virus's (EBV's) latent-lytic switch BZLF1 gene, Zp, inducing viral reactivation. In this study, EBV-infected cell lines derived from gastric cancers and Burkitt lymphomas were incubated with HIF-1α-stabilizing drugs: the iron chelator deferoxamine (Desferal [DFO]), a neddylation inhibitor (pevonedistat [MLN-4924]), and a prolyl hydroxylase inhibitor (roxadustat [FG-4592]). DFO and MLN-4924, but not FG-4592, induced accumulation of both lytic EBV proteins and phosphorylated p53 in cell lines that contain a wild-type p53 gene. FG-4592 also failed to activate transcription from Zp in a reporter assay despite inducing accumulation of HIF-1α and transcription from another HRE-containing promoter. Unexpectedly, DFO failed to induce EBV reactivation in cell lines that express mutant or no p53 or when p53 expression was knocked down with short hairpin RNAs (shRNAs). Likewise, HIF-1α failed to activate transcription from Zp when p53 was knocked out by CRISPR-Cas9. Importantly, DFO induced binding of p53 as well as HIF-1α to Zp in chromatin immunoprecipitation (ChIP) assays, but only when the HRE was present. Nutlin-3, a drug known to induce accumulation of phosphorylated p53, synergized with DFO and MLN-4924 in inducing EBV reactivation. Conversely, KU-55933, a drug that inhibits ataxia telangiectasia mutated, thereby preventing p53 phosphorylation, inhibited DFO-induced EBV reactivation. Lastly, activation of Zp transcription by DFO and MLN-4924 mapped to its HRE. Thus, we conclude that induction of BZLF1 gene expression by HIF-1α requires phosphorylated, wild-type p53 as a coactivator, with HIF-1α binding recruiting p53 to Zp.IMPORTANCE EBV, a human herpesvirus, is latently present in most nasopharyngeal carcinomas, Burkitt lymphomas, and some gastric cancers. To develop a lytic-induction therapy for treating patients with EBV-associated cancers, we need a way to efficiently reactivate EBV into lytic replication. EBV's BZLF1 gene product, Zta, usually controls this reactivation switch. We previously showed that HIF-1α binds the BZLF1 gene promoter, inducing Zta synthesis, and HIF-1α-stabilizing drugs can induce EBV reactivation. In this study, we determined which EBV-positive cell lines are reactivated by classes of HIF-1α-stabilizing drugs. We found, unexpectedly, that HIF-1α-stabilizing drugs only induce reactivation when they also induce accumulation of phosphorylated, wild-type p53. Fortunately, p53 phosphorylation can also be provided by drugs such as nutlin-3, leading to synergistic reactivation of EBV. These findings indicate that some HIF-1α-stabilizing drugs may be helpful as part of a lytic-induction therapy for treating patients with EBV-positive malignancies that contain wild-type p53.
Collapse
MESH Headings
- Cell Line, Tumor
- Cyclopentanes/pharmacology
- Deferoxamine/pharmacology
- Enzyme Inhibitors/pharmacology
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/virology
- Gene Expression Regulation
- Glycine/analogs & derivatives
- Glycine/pharmacology
- Herpesvirus 4, Human/drug effects
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/growth & development
- Herpesvirus 4, Human/metabolism
- Host-Pathogen Interactions/drug effects
- Host-Pathogen Interactions/genetics
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/agonists
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Imidazoles/pharmacology
- Iron Chelating Agents/pharmacology
- Isoquinolines/pharmacology
- Lymphocytes/drug effects
- Lymphocytes/metabolism
- Lymphocytes/virology
- Morpholines/pharmacology
- Piperazines/pharmacology
- Prolyl-Hydroxylase Inhibitors/pharmacology
- Promoter Regions, Genetic
- Protein Binding/drug effects
- Pyrimidines/pharmacology
- Pyrones/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Response Elements
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Tumor Suppressor Protein p53/antagonists & inhibitors
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Virus Activation/drug effects
Collapse
Affiliation(s)
- Richard J Kraus
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Blue-Leaf A Cordes
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Saraniya Sathiamoorthi
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Parita Patel
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Xueying Yuan
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tawin Iempridee
- National Nanotechnology Center, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Xianming Yu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Denis L Lee
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Janet E Mertz
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
91
|
Jiang J, Zhang X, Tang Y, Li S, Chen J. Progress on ocular siRNA gene-silencing therapy and drug delivery systems. Fundam Clin Pharmacol 2020; 35:4-24. [PMID: 32298491 DOI: 10.1111/fcp.12561] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/24/2020] [Accepted: 04/10/2020] [Indexed: 12/14/2022]
Abstract
Age-related macular degeneration (AMD) and glaucoma are global ocular diseases with high blindness rate. RNA interference (RNAi) is being increasingly used in the treatment of these disorders with siRNA drugs, bevasiranib, AGN211745 and PF-04523655 for AMD, and SYL040012 and QPI-1007 for glaucoma. Administration routes and vectors of gene drugs affect their therapeutic effect. Compared with the non-viral vectors, viral vectors have limited payload capacity and potential immunogenicity. This review summarizes the progress of the ocular siRNA gene-silencing therapy by focusing on siRNA drugs for AMD and glaucoma already used in clinical research, the main routes of drug delivery and the non-viral vectors for siRNA drugs.
Collapse
Affiliation(s)
- Jinjin Jiang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.1, DongQing Road, Guiyang, 550014, People's Republic of China.,Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing, 211198, People's Republic of China
| | - Xinru Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.1, DongQing Road, Guiyang, 550014, People's Republic of China.,Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing, 211198, People's Republic of China
| | - Yue Tang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.1, DongQing Road, Guiyang, 550014, People's Republic of China.,Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing, 211198, People's Republic of China
| | - Shuhan Li
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing, 211198, People's Republic of China
| | - Jing Chen
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing, 211198, People's Republic of China
| |
Collapse
|
92
|
Priya Dharshini LC, Vishnupriya S, Sakthivel KM, Rasmi RR. Oxidative stress responsive transcription factors in cellular signalling transduction mechanisms. Cell Signal 2020; 72:109670. [PMID: 32418887 DOI: 10.1016/j.cellsig.2020.109670] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 11/18/2022]
Abstract
Oxidative stress results from the imbalances in the development of reactive oxygen species (ROS) and antioxidants defence system resulting in tissue injury. A key issue resulting in the modulation of ROS is that it alters hosts molecular, structural and functional properties which is accomplished via various signalling pathways which either activate or inhibit numerous transcription factors (TFs). Some of the regulators include Nuclear erythroid-2 related factors (Nrf-2), CCAAT/enhancer-binding protein delta (CEBPD), Activator Protein-1 (AP-1), Hypoxia-inducible factor 1(HIF-1), Nuclear factor κB (NF-κB), Specificity Protein-1 (SP-1) and Forkhead Box class O (FoxO) transcription factors. The expression of these transcription factors are dependent upon the stress signal and are sometimes interlinked. They are highly specific having their own regulation cellular events. Depending upon the transcription factors and better knowledge on the type of the oxidative stress help researchers develop safe, novel targets which can serve as efficient therapeutic targets for several disease conditions.
Collapse
Affiliation(s)
| | - Selvaraj Vishnupriya
- Department of Biotechnology, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India
| | - Kunnathur Murugesan Sakthivel
- Department of Biochemistry, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India
| | - Rajan Radha Rasmi
- Department of Biotechnology, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India.
| |
Collapse
|
93
|
Schwenzer H, Jühling F, Chu A, Pallett LJ, Baumert TF, Maini M, Fassati A. Oxidative Stress Triggers Selective tRNA Retrograde Transport in Human Cells during the Integrated Stress Response. Cell Rep 2020; 26:3416-3428.e5. [PMID: 30893612 PMCID: PMC6426654 DOI: 10.1016/j.celrep.2019.02.077] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 01/05/2023] Open
Abstract
In eukaryotes, tRNAs are transcribed in the nucleus and exported to the cytosol, where they deliver amino acids to ribosomes for protein translation. This nuclear-cytoplasmic movement was believed to be unidirectional. However, active shuttling of tRNAs, named tRNA retrograde transport, between the cytosol and nucleus has been discovered. This pathway is conserved in eukaryotes, suggesting a fundamental function; however, little is known about its role in human cells. Here we report that, in human cells, oxidative stress triggers tRNA retrograde transport, which is rapid, reversible, and selective for certain tRNA species, mostly with shorter 3′ ends. Retrograde transport of tRNASeC, which promotes translation of selenoproteins required to maintain homeostatic redox levels in cells, is highly efficient. tRNA retrograde transport is regulated by the integrated stress response pathway via the PERK-REDD1-mTOR axis. Thus, we propose that tRNA retrograde transport is part of the cellular response to oxidative stress. Oxidative stress triggers nuclear import of cytoplasmic tRNAs Import is selective for certain tRNAs Import requires activation of the unfolded protein response and inhibition of mTOR via REDD1 tRNA nuclear import is a component of the integrated stress response
Collapse
Affiliation(s)
- Hagen Schwenzer
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Frank Jühling
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 2 Université de Strasbourg, 67000 Strasbourg, France
| | - Alexander Chu
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Laura J Pallett
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Thomas F Baumert
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 2 Université de Strasbourg, 67000 Strasbourg, France; Nouvel Hôpital Civil, Institut Hospitalo-Universitaire, 67000 Strasbourg, France
| | - Mala Maini
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Ariberto Fassati
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK.
| |
Collapse
|
94
|
Vishlaghi N, Lisse TS. Exploring vitamin D signalling within skin cancer. Clin Endocrinol (Oxf) 2020; 92:273-281. [PMID: 31889334 DOI: 10.1111/cen.14150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
Abstract
Sunlight exposure of the skin is associated with both risks and benefits. On one hand, sunlight ultraviolet (UV) radiation can cause skin cancer through signature DNA mutations. On the other hand, it can be absorbed in the skin by 7-dehydrocholesterol to instigate endogenous synthesis of vitamin D to regulate anticancer effects. Thus, protecting one's skin from sunlight to avoid skin cancer may lead to impaired vitamin D levels arguing for sensible sun exposure practices. To limit cancer, vitamin D metabolites can promote uncharacterized and diverse sets of events such as repair responses to DNA damage, apoptosis of malignant cells, and suppression of immune surveillance, proliferation and angiogenesis. Recent findings also suggest that part of the anticancer effects of vitamin D within squamous cell carcinoma-a type of skin cancer most directly linked to sun exposure-involves the DDIT4-mTOR catabolic signalling pathway to enhance cell autophagy. As mTOR activity and cellular metabolism are modulated as part of the DNA damage response, insights into the means by which mTOR can be controlled by vitamin D to suppress cancer is of molecular and clinical importance. Overall, the research so far suggests that presence of vitamin D through sunlight exposure and supplementation are beneficial for human health in the face of cancer.
Collapse
Affiliation(s)
- Neda Vishlaghi
- Cox Science Center, Biology Department, University of Miami, Coral Gables, FL, USA
| | - Thomas S Lisse
- Cox Science Center, Biology Department, University of Miami, Coral Gables, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
95
|
Dumas K, Ayachi C, Gilleron J, Lacas‐Gervais S, Pastor F, Favier FB, Peraldi P, Vaillant N, Yvan‐Charvet L, Bonnafous S, Patouraux S, Anty R, Tran A, Gual P, Cormont M, Tanti J, Giorgetti‐Peraldi S. REDD1 deficiency protects against nonalcoholic hepatic steatosis induced by high‐fat diet. FASEB J 2020; 34:5046-5060. [DOI: 10.1096/fj.201901799rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/24/2020] [Accepted: 01/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Karine Dumas
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Chaima Ayachi
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Jerome Gilleron
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | | | - Faustine Pastor
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | | | - Pascal Peraldi
- Université Côte d’Azur, Inserm, CNRS, iBV, Team “Stem Cells and Differentiation” France
| | - Nathalie Vaillant
- Université Côte d’Azur, Inserm, C3M, Team “Haematometabolism in Diseases” France
| | - Laurent Yvan‐Charvet
- Université Côte d’Azur, Inserm, C3M, Team “Haematometabolism in Diseases” France
| | - Stéphanie Bonnafous
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Stéphanie Patouraux
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Rodolphe Anty
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Albert Tran
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
- Université Côte d’Azur, CHU, Inserm, C3M,Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Philippe Gual
- Université Côte d’Azur, Inserm, C3M, Team “Chronic Liver Diseases Associated with Steatosis and Alcohol” France
| | - Mireille Cormont
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Jean‐François Tanti
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| | - Sophie Giorgetti‐Peraldi
- Université Côte d’Azur, Inserm, C3M, Team “Cellular and Molecular Physiopathology of Obesity” France
| |
Collapse
|
96
|
Shen-Hui X, Fu WW, Zhang J, Wang HP, Dang K, Chang H, Gao YF. Different fuel regulation in two types of myofiber results in different antioxidant strategies in Daurian ground squirrels (Spermophilus dauricus) during hibernation. J Exp Biol 2020:jeb.231639. [PMID: 34005794 DOI: 10.1242/jeb.231639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 12/08/2020] [Indexed: 11/20/2022]
Abstract
We previously showed that different skeletal muscles in Daurian ground squirrels (Spermophilus dauricus) possess different antioxidant strategies during hibernation; however, the reason for these varied strategies remains unclear. To clarify this issue, we studied REDD1, FOXO4, PGC-1α, FOXO1, and atrogin-1 proteins to determine the potential cause of the different antioxidant strategies in Daurian ground squirrels during hibernation, and to clarify whether different strategies affect atrophy-related signals. Results showed that the soleus (SOL) muscle experienced intracellular hypoxia during interbout arousal, but no oxidative stress. This may be due to increased PGC-1α expression enhancing antioxidant capacity in the SOL under hypoxic conditions. Extensor digitorum longus (EDL) muscle showed no change in oxidative stress, hypoxia, or antioxidant capacity during hibernation. The FOXO1 and PGC-1α results strongly suggested differentially regulated fuel metabolism in the SOL and EDL muscles during hibernation, i.e., enhanced lipid oxidation and maintained anaerobic glycolysis, respectively. Atrogin-1 expression did not increase during hibernation in either the SOL or EDL, indicating that protein synthesis was not inhibited by atrogin-1. Thus, our results suggest that different fuel regulation may be one mechanism related to antioxidant defense strategy formation in different kinds of skeletal muscle fibers of Daurian ground squirrels during hibernation.
Collapse
Affiliation(s)
- Xu Shen-Hui
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Wei-Wei Fu
- Shaanxi Key Laboratory for Animal Conservation, Shaanxi Institute of Zoology, Xi'an Shaanxi 710032, China
| | - Jie Zhang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Hui-Ping Wang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Kai Dang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an Shaanxi, China
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| | - Yun-Fang Gao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an Shaanxi 710069, China
| |
Collapse
|
97
|
Feng Y, Song K, Shang W, Chen L, Wang C, Pang B, Wang N. REDD1 overexpression in oral squamous cell carcinoma may predict poor prognosis and correlates with high microvessel density. Oncol Lett 2020; 19:431-441. [PMID: 31897156 PMCID: PMC6923876 DOI: 10.3892/ol.2019.11070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/11/2019] [Indexed: 01/10/2023] Open
Abstract
The association between the hypoxia-inducible gene termed regulated in development and DNA damage responses 1 (REDD1) and microvessel density (MVD) in human oral cancer has rarely been reported. The present study aimed to explore REDD1 expression in oral squamous cell carcinoma (OSCC), its clinical prognostic significance and its correlation with angiogenesis. REDD1 expression in 23 pairs of fresh-frozen OSCC and matched peritumoral mucosal tissues was quantified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. Furthermore, 74 formalin-fixed paraffin-embedded OSCC tissues were collected to detect REDD1 expression and CD34-positive MVD by immunohistochemistry (IHC). The association between REDD1 expression and MVD, patients' clinicopathological characteristics and cancer-associated survival rate was also evaluated using the log-rank (Mantel-Cox) test. The results from RT-qPCR and western blotting demonstrated that REDD1 expression was significantly higher in OSCC tissues compared with peritumoral mucosal tissues (P<0.05). In addition, the results from IHC revealed that REDD1 expression was higher in OSCC tissues compared with peritumoral tissues. Furthermore, REDD1 expression was associated with advanced clinical stage, poorer tumor differentiation, lymphatic metastasis and tumor recurrence (P=0.000, P=0.003, P=0.006 and P<0.001, respectively). Additionally, REDD1 overexpression was positively correlated with MVD (r=0.7316; P<0.001). The results from Kaplan-Meier survival analysis demonstrated a significantly reduced disease-free survival and overall survival in patients with OSCC and high REDD1 expression (P<0.001). REDD1 may therefore serve as a novel prognostic biomarker, a key regulatory checkpoint that could coordinate angiogenesis and a new therapeutic target for patients with OSCC.
Collapse
Affiliation(s)
- Yuanyong Feng
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Kai Song
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wei Shang
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Liqiang Chen
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Chengqin Wang
- Department of Pathology, Basic Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Baoxing Pang
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ning Wang
- Department of Pathology, Basic Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
98
|
Protective Role of mTOR in Liver Ischemia/Reperfusion Injury: Involvement of Inflammation and Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7861290. [PMID: 31827701 PMCID: PMC6885218 DOI: 10.1155/2019/7861290] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/24/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Liver ischemia/reperfusion (IR) injury is a common phenomenon after liver resection and transplantation, which often results in liver graft dysfunction such as delayed graft function and primary nonfunction. The mammalian target of rapamycin (mTOR) is an evolutionarily highly conserved serine/threonine protein kinase, which coordinates cell growth and metabolism through sensing environmental inputs under physiological or pathological conditions, involved in the pathophysiological process of IR injury. In this review, we mainly present current evidence of the beneficial role of mTOR in modulating inflammation and autophagy under liver IR to provide some evidence for the potential therapies for liver IR injury.
Collapse
|
99
|
Miller WP, Toro AL, Barber AJ, Dennis MD. REDD1 Activates a ROS-Generating Feedback Loop in the Retina of Diabetic Mice. Invest Ophthalmol Vis Sci 2019; 60:2369-2379. [PMID: 31141608 PMCID: PMC6541429 DOI: 10.1167/iovs.19-26606] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The present study was designed to evaluate the role of the stress response protein REDD1 in diabetes-induced oxidative stress and retinal pathology. Methods Wild-type and REDD1-deficient mice were administered streptozotocin to induce diabetes. Some mice received the antioxidant N-acetyl-l-cysteine (NAC). Visual function was assessed by virtual optometry. Retinas were analyzed by Western blotting. Reactive oxygen species (ROS) were assessed by 2,7-dichlorofluoroscein. Similar analyses were performed on R28 retinal cells in culture exposed to hyperglycemic conditions, NAC, and/or the exogenous ROS source hydrogen peroxide. Results In the retina of diabetic mice, REDD1 expression and ROS were increased. In cells in culture, hyperglycemic conditions enhanced REDD1 expression, ROS levels, and the mitochondrial membrane potential. However, similar effects were not observed in the retina of diabetic mice or cells lacking REDD1. In the retina of diabetic mice and cells exposed to hyperglycemic conditions, NAC normalized ROS and prevented an increase in REDD1 expression. Diabetic mice receiving NAC also exhibited improved contrast sensitivity as compared to diabetic controls. Hydrogen peroxide addition to culture medium increased REDD1 expression and attenuated Akt/GSK3 phosphorylation in a REDD1-dependent manner. In REDD1-deficient cells exposed to hyperglycemic conditions, expression of a dominant negative Akt or constitutively active GSK3 increased the mitochondrial membrane potential and promoted ROS. Conclusions The findings provide new insight into the mechanism whereby diabetes-induced hyperglycemia causes oxidative stress and visual dysfunction. Specifically, hyperglycemia-induced REDD1 activates a ROS-generating feedback loop that includes Akt/GSK3. Thus, therapeutic approaches targeting REDD1 expression and ROS may be beneficial for preventing diabetes-induced visual dysfunction.
Collapse
Affiliation(s)
- William P Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Allyson L Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Alistair J Barber
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States.,Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States.,Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, United States
| |
Collapse
|
100
|
Li P, Lin N, Guo M, Huang H, Yu T, Zhang L. REDD1 knockdown protects H9c2 cells against myocardial ischemia/reperfusion injury through Akt/mTORC1/Nrf2 pathway-ameliorated oxidative stress: An in vitro study. Biochem Biophys Res Commun 2019; 519:179-185. [DOI: 10.1016/j.bbrc.2019.08.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/16/2019] [Indexed: 01/06/2023]
|