1
|
Bose GS, Kalakoti G, Kulkarni AP, Mittal S. AP-1/C-FOS and AP-1/FRA2 differentially regulate early and late adipogenic differentiation of mesenchymal stem cells. J Cell Biochem 2024; 125:e30543. [PMID: 38440920 DOI: 10.1002/jcb.30543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 03/06/2024]
Abstract
Obesity is defined as an abnormal accumulation of adipose tissue in the body and is a major global health problem due to increased morbidity and mortality. Adipose tissue is made up of adipocytes, which are fat-storing cells, and the differentiation of these fat cells is known as adipogenesis. Several transcription factors (TFs) such as CEBPβ, CEBPα, PPARγ, GATA, and KLF have been reported to play a key role in adipogenesis. In this study, we report one more TF AP-1, which is found to be involved in adipogenesis. Human mesenchymal stem cells were differentiated into adipocytes, and the expression pattern of different subunits of AP-1 was examined during adipogenesis. It was observed that C-FOS was predominantly expressed at an early stage (Day 2), whereas FRA2 expression peaked at later stages (Days 6 and 8) of adipogenesis. Chromatin immunoprecipitation-sequencing analysis revealed that C-FOS binds mainly to the promoters of WNT1, miR-30a, and ANAPC7 and regulates their expression during mitotic clonal expansion. In contrast, FRA2 binds to the promoters of CIDEA, NOTCH1, ARAF, and MYLK, regulating their expression and lipid metabolism. Data obtained clearly indicate that the differential expression of C-FOS and FRA2 is crucial for different stages of adipogenesis. This also raises the possibility of considering AP-1 as a therapeutic target for treating obesity and related disorders.
Collapse
Affiliation(s)
- Ganesh Suraj Bose
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Garima Kalakoti
- Bioinformatics Center, Savitribai Phule Pune University, Pune, India
| | | | - Smriti Mittal
- Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
2
|
Liu T, Xia S. The Proteostasis of Thymic Stromal Cells in Health and Diseases. Protein J 2024; 43:447-463. [PMID: 38622349 DOI: 10.1007/s10930-024-10197-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
The thymus is the key immune organ for the development of T cells. Different populations of thymic stromal cells interact with T cells, thereby controlling the dynamic development of T cells through their differentiation and function. Proteostasis represents a balance between protein expression, folding, and modification and protein clearance, and its fluctuation usually depends at least partially on related protein regulatory systems for further survival and effects. However, in terms of the substantial requirement for self-antigens and their processing burden, increasing evidence highlights that protein regulation contributes to the physiological effects of thymic stromal cells. Impaired proteostasis may expedite the progression of thymic involution and dysfunction, accompanied by the development of autoimmune diseases or thymoma. Hence, in this review, we summarize the regulation of proteostasis within different types of thymic stromal cells under physiological and pathological conditions to identify potential targets for thymic regeneration and immunotherapy.
Collapse
Affiliation(s)
- Ting Liu
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
3
|
Wang Z, Su Y, Zhao M, Ma Z, Li J, Hou Z, Li H. NOTCH1 as a Negative Regulator of Avian Adipocyte Differentiation: Implications for Fat Deposition. Animals (Basel) 2024; 14:585. [PMID: 38396553 PMCID: PMC10886207 DOI: 10.3390/ani14040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The NOTCH signaling pathway plays a pivotal role in diverse developmental processes, including cell proliferation and differentiation. In this study, we investigated whether this signaling molecules also contribute to avian adipogenesis. Using previous mRNA-seq datasets, we examined the expression of 11 signaling members during avian adipocyte differentiation. We found most members are down-regulated throughout differentiation (p < 0.05). As a representative, NOTCH1 was decreased in cultured chicken abdominal adipocytes during adipogenesis at mRNA and protein levels (p < 0.05). Moreover, using an overexpression plasmid for NOTCH1's intracellular domain (NICD1), as well as siRNA and DAPT to activate or deplete NOTCH1 in cells, we investigated the role of NOTCH1 in avian adipogenesis. Our findings illuminate that NOTCH1 activates the expression of HES1 and SOCS3 while it decreases NR2F2 and NUMB (p < 0.05), as well as inhibits oleic acid-induced adipocyte differentiation (p < 0.01). We further demonstrate that HES1, a downstream transcription factor activated by NOTCH1, also significantly inhibits adipogenesis by suppressing PPARγ and C/EBPα (p < 0.01). Collectively, these findings establish NOTCH1 as a negative regulator of avian adipocyte differentiation, unveiling NOTCH signaling as a potential target for regulating avian fat deposition.
Collapse
Affiliation(s)
- Zheng Wang
- College of Life Sciences, Shanxi Agricultural University, Jinzhong 030801, China; (Z.W.); (Y.S.); (M.Z.); (Z.M.)
| | - Yue Su
- College of Life Sciences, Shanxi Agricultural University, Jinzhong 030801, China; (Z.W.); (Y.S.); (M.Z.); (Z.M.)
| | - Mingyu Zhao
- College of Life Sciences, Shanxi Agricultural University, Jinzhong 030801, China; (Z.W.); (Y.S.); (M.Z.); (Z.M.)
| | - Zhenhua Ma
- College of Life Sciences, Shanxi Agricultural University, Jinzhong 030801, China; (Z.W.); (Y.S.); (M.Z.); (Z.M.)
| | - Jianhui Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China;
| | - Zhuocheng Hou
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Huifeng Li
- College of Life Sciences, Shanxi Agricultural University, Jinzhong 030801, China; (Z.W.); (Y.S.); (M.Z.); (Z.M.)
| |
Collapse
|
4
|
Wang Z, Li Y, Wu L, Guo Y, Yang G, Li X, Shi X. Rosiglitazone-induced PPARγ activation promotes intramuscular adipocyte adipogenesis of pig. Anim Biotechnol 2023; 34:3708-3717. [PMID: 37149785 DOI: 10.1080/10495398.2023.2206872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Intramuscular fat (IMF) positively influences various aspects of meat quality, while the subcutaneous fat (SF) has negative effect on carcass characteristics and fattening efficiency. Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of adipocyte differentiation, herein, through bioinformatic screen for the potential regulators of adipogenesis from two independent microarray datasets, we identified that PPARγ is a potentially regulator between porcine IMF and SF adipogenesis. Then we treated subcutaneous preadipocytes (SA) and intramuscular preadipocytes (IMA) of pig with RSG (1 µmol/L), and we found that RSG treatment promoted the differentiation of IMA via differentially activating PPARγ transcriptional activity. Besides, RSG treatment promoted apoptosis and lipolysis of SA. Meanwhile, by the treatment of conditioned medium, we excluded the possibility of indirect regulation of RSG from myocyte to adipocyte and proposed that AMPK may mediate the RSG-induced differential activation of PPARγ. Collectively, the RSG treatment promotes IMA adipogenesis, and advances SA lipolysis, this effect may be associated with AMPK-mediated PPARγ differential activation. Our data indicates that targeting PPARγ might be an effective strategy to promote intramuscular fat deposition while reduce subcutaneous fat mass of pig.
Collapse
Affiliation(s)
- Zhaolu Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Youlei Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Lingling Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Yuan Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Xiao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| | - Xin'e Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A & F University, Shaanxi, P. R. China
| |
Collapse
|
5
|
Osathanon T, Egusa H. Notch signaling in induced pluripotent stem cells. MOLECULAR PLAYERS IN IPSC TECHNOLOGY 2022:249-284. [DOI: 10.1016/b978-0-323-90059-1.00003-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
6
|
Du H, Wang Y, Liu X, Wang S, Wu S, Yuan Z, Zhu X. miRNA-146a-5p mitigates stress-induced premature senescence of D-galactose-induced primary thymic stromal cells. Cytokine 2021; 137:155314. [PMID: 33002743 DOI: 10.1016/j.cyto.2020.155314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Senescent thymic stromal cells (TSCs) producing senescence-associated secretory phenotype (SASP) may play a role at later phases of thymic involution. However, the etiology and mechanisms responsible for TSC senescence remain to be elucidated. In the present study, the effects of oxidative stress on TSCs and role of miRNA-146a-5p in stress-induced premature senescence (SIPS) were identified. D-galactose (D-gal) induced oxidative stress in primary TSCs and a limited cumulative oxidative stress induced premature senescence but not apoptosis of TSCs. miRNA-146a-5p overexpression can mitigate the SIPS by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6) instead of increasing autophagy clearance. Furthermore, exogenous miRNA-146a-5p reversed the upregulation of chemokines including Cxcl5, pro-inflammatory cytokines, and antimicrobial peptides in TSCs with SIPS. In conclusion, the accumulated oxidative stress may be partially responsible for senescence in TSCs and modulation of miRNA-146a-5p may attenuate this process.
Collapse
Affiliation(s)
- Hongmei Du
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China; Department of Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Yajun Wang
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China
| | - Xue Liu
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Simeng Wu
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Zhe Yuan
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, 39 Huaxiang Road, Shenyang, China
| | - Xike Zhu
- Research Center, Shengjing Hospital of China Medical University, 7 Mulan Road, Economic Development Zone, Benxi, China.
| |
Collapse
|
7
|
Liu MC, Logan H, Newman JJ. Distinct roles for Notch1 and Notch3 in human adipose-derived stem/stromal cell adipogenesis. Mol Biol Rep 2020; 47:8439-8450. [PMID: 33021719 DOI: 10.1007/s11033-020-05884-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
The role of the Notch signaling pathway in adipogenesis has long been controversial as the action of individual Notch receptors appears to vary with experimental conditions. In this study, we offer some explanation for the observed contradictions by comparing the role of both Notch1 and Notch3 in regulating the expression of key adipogenic regulator, PPARγ, in human adipose-derived stem/stromal cells (hADSCs) during in vitro adipogenesis. Utilizing qRT-PCR, western blot, and immunofluorescence staining, we demonstrated that Notch3 was expressed prior to the formation of lipid vesicles, while Notch1 only appeared after vesicle formation. In addition, following the induction of adipogenesis, the levels of Notch1 intracellular domain in the nucleus were significantly reduced, while the siRNA-mediated loss of Notch1 reduced transcript but not protein levels of PPARγ. The knockdown of Notch3 led to increased expression of PPARγ during early adipogenesis that was not paralleled by a decreased expression of Hes1 and Hey1, but was accompanied by a marked decrease in the protein level of β-catenin, the key functional component of the canonical Wnt/β-catenin signaling pathway. This study deepens the understanding of the Notch pathway by clarifying the distinct roles of Notch1 and Notch3 during adipogenesis. We showed that Notch3 is involved in early adipogenic differentiation, while Notch1 functions later in the process. In addition, we begin to uncover the interaction between the Notch and Wnt signaling pathways that may offer novel therapeutic targets aimed at obesity and diabetes.
Collapse
Affiliation(s)
- Meng-Cheng Liu
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Hannah Logan
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Jamie J Newman
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, 71272, USA.
| |
Collapse
|
8
|
Koutroulis I, Batabyal R, McNamara B, Ledda M, Hoptay C, Freishtat RJ. Sepsis Immunometabolism: From Defining Sepsis to Understanding How Energy Production Affects Immune Response. Crit Care Explor 2019; 1:e0061. [PMID: 32166242 PMCID: PMC7063962 DOI: 10.1097/cce.0000000000000061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES This review will examine current definitions and trends in sepsis management as well pathophysiologic mechanisms in animal and ex vivo studies that correlate decreased energy production with deranged inflammatory response during the septic process. DATA SOURCES The latest articles in the literature that focus on the role of immunometabolism and associated mechanisms in sepsis were selected. STUDY SELECTION The most relevant, original articles were included in the review. DATA EXTRACTION All pertinent data for sepsis definitions as well as changes in immunometabolic pathways during the septic process was reviewed and assessed for inclusion in this article. DATA SYNTHESIS Sepsis is a major cause of multiple organ dysfunction. It is the principal cause of death resulting from infection and one of the most expensive conditions treated in the United States. Despite current efforts to accurately define sepsis, novel treatments and highly trained providers, mortality rates for sepsis remain high, prompting a need for further investigation of underlying immunometabolic mechanisms to identify potential treatment targets. The definition of sepsis has shifted and changed in the past few decades due to poorly defined criteria, as well as unclear guidelines for providers with regards to management of severe sepsis and septic shock. The early identification of patients with a systemic inflammatory response that will progress to septic shock is critical since recent traditional therapeutic approaches, such as early goal-directed therapy, IV immunoglobulin, and anti-tumor necrosis factor-α antibodies have failed. CONCLUSIONS There are no effective anti-sepsis drug therapies due to complex inflammatory and metabolic interactions. Further studies regarding the interface between innate immunity and metabolism should be investigated to effectively address septic patient mortality rates.
Collapse
Affiliation(s)
- Ioannis Koutroulis
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Rachael Batabyal
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Brittany McNamara
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Matthew Ledda
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
| | - Claire Hoptay
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Robert J Freishtat
- Division of Emergency Medicine, Children's National Hospital, Washington, DC
- Center for Genetic Medicine, Children's National Research Institute, Washington, DC
- George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
9
|
Sun LJ, Qiao W, Xiao YJ, Cui L, Wang X, Ren WD. Naringin mitigates myocardial strain and the inflammatory response in sepsis-induced myocardial dysfunction through regulation of PI3K/AKT/NF-κB pathway. Int Immunopharmacol 2019; 75:105782. [PMID: 31376623 DOI: 10.1016/j.intimp.2019.105782] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/18/2019] [Accepted: 07/24/2019] [Indexed: 12/27/2022]
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is a manifestation of severe sepsis and is the main cause of increased mortality in sepsis patients. Naringin (Nar) has been reported to possess various biological activities and pharmacological properties. Therefore, the present study was undertaken to evaluate whether Nar can protect rats from the effects of LPS-induced SIMD. SD Rats were pre-treated with Nar (50 and 100 mg/kg) for 7 days before administration of a single dose of LPS (10 mg/kg, i.p.) on the seventh day. We found that Nar treatment markedly improved the global strain and strain rate of longitudinal, circumference, and radial direction (GLS/GLSr, GCS/GCSr, GRS/GRSr) compared to the LPS group. The layer-specific strain decreased gradually from the endocardial layer to epicardial layer, and the most serious damage occurred in the endocardial layer. Moreover, Nar significantly decreased the levels of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and myocardial enzymes (CK, LDH, and AST) induced by LPS and attenuated the inflammation response. Finally, Nar also inhibited NF-κB nuclear translocation and the activity of iNOS in H9c2 cardiomyocytes by activating PI3K/AKT signaling pathway. These results suggest that naringin may possess novel therapeutic potential for protection against LPS-induced myocardial dysfunction.
Collapse
Affiliation(s)
- Li-Juan Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Ultrasound, The First Hospital of Qinhuangdao, Qinhuangdao 066000, PR China
| | - Wei Qiao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Yang-Jie Xiao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Li Cui
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Xin Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Wei-Dong Ren
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
10
|
Tan J, Wang Y, Wang S, Wu S, Yuan Z, Zhu X. Label-free quantitative proteomics identifies transforming growth factor β1 (TGF-β1) as an inhibitor of adipogenic transformation in OP9-DL1 cells and primary thymic stromal cells. Cell Biosci 2019; 9:48. [PMID: 31249661 PMCID: PMC6570845 DOI: 10.1186/s13578-019-0311-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/08/2019] [Indexed: 11/21/2022] Open
Abstract
Background Adipocyte accumulation is a predominant feature of age-related thymic involution, but the mechanisms responsible for thymic adipogenesis remain to be elucidated. The aim of this study was to identify key regulators in thymic adipogenesis. We used rosiglitazone, a potent peroxisome proliferator-activated receptor γ (PPARγ) agonist, to induce adipogenic differentiation of OP9-DL1 cells, and investigated the differentially expressed proteins during adipogenic differentiation by using label-free quantitative proteomics. Furthermore, the effects of transforming growth factor β1 (TGF-β1) on rosiglitazone-induced adipogenic differentiation of OP9-DL1 cells as well as the underlying mechanisms were also investigated. Results Proteomic analysis identified 139 proteins differed significantly in rosiglitazone-treated cells compared with dimethyl sulphoxide (DMSO)-treated cells. Rosiglitazone-induced adipogenic differentiation was inhibited by TGF-β1 treatment in OP9-DL1 cells and primary thymic stromal cells. Real-time PCR analysis showed significant increases in PPARγ and fatty acid binding protein 4 mRNA levels in rosiglitazone-treated cells, which were inhibited by TGF-β1 treatment. TGF-β1 down-regulated PPARγ expression at both mRNA and protein levels in OP9-DL1 cells. Chromatin immunoprecipitation analysis demonstrated that TGF-β1 enhanced the binding of Smad2/3 and histone deacetylase 1, but reduced the binding of H3K14ac to the promoter of PPARγ gene. TGF-β1 partially reversed the inhibitory effects of rosiglitazone on the expression of Axin2 and β-catenin protein levels. TGF-β1 inhibited rosiglitazone-induced adipogenic transformation in OP9-DL1 cells by down-regulation of PPARγ and activation of the canonical Wnt/β-catenin signaling pathway. Conclusion Taken together, activation of TGF-β pathway may serve as a useful strategy to prevent thymic adiposity in age-related thymic involution. Electronic supplementary material The online version of this article (10.1186/s13578-019-0311-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianxin Tan
- 1State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004 People's Republic of China
| | - Yajun Wang
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| | - Siliang Wang
- 3Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Simeng Wu
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Zhe Yuan
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Xike Zhu
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| |
Collapse
|
11
|
A novel role for NFIA in restoring radiosensitivity in radioresistant NSCLC cells by downregulating the AKT and ERK pathways. Biochem Biophys Res Commun 2019; 515:558-564. [PMID: 31178144 DOI: 10.1016/j.bbrc.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 12/27/2022]
Abstract
Radioresistance remains the most challenging issue leading to radiotherapy failure in the treatment of non-small cell lung cancer (NSCLC). The nuclear factor IA (NFIA) is associated with tumor response to treatments in many cancers, but its role in NSCLC radioresistance remains unclear. Here, we established two radioresistant NSCLC cell lines, H226R and H460R, by dose-gradient irradiation to investigate the function of NFIA in NSCLC radioresistance. The results showed a dramatically reduced expression of NFIA in radioresistant cells accompanied with elevated phosphorylation of AKT and ERK, when compared with their parental cells. Overexpression of NFIA restored the sensitivity of radioresistant cells to radiation through increased ionizing radiation (IR)-induced apoptosis and DNA damage by downregulating p-AKT and p-ERK, whereas knockdown of NFIA promoted radioresistance of the parental cells. Our findings suggested that NFIA enhanced cell radiosensitivity by downregulating p-AKT and p-ERK in NSCLC. Our study fills a gap in the field of NFIA and radioresistance, and establishes a mechanistic foundation to improve radiotherapy efficiency in NSCLC patients.
Collapse
|