1
|
Abudureyimu S, He C, Xie W, Chen Z, Airikenjiang H, Abulaiti D, Cao Y, Qiu H, Gao Y. FOXO3a functions as a transcriptional and co-transcriptional splicing regulator in vascular endothelial cell lines. Gene 2024; 904:148221. [PMID: 38286271 DOI: 10.1016/j.gene.2024.148221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024]
Abstract
Recent studies have indicated a connection between Forkhead box O3a protein and coronary artery disease, yet the exact role of FOXO3a in the regulation of metabolic processes and apoptosis in vascular endothelial cells is still unknown. Therefore, we investigated the role of FOXO3a on target genes in a human vascular endothelial cell line. Through the utilization of high-throughput sequencing technology, we analyzed gene expression profiles and alternative splicing patterns in human vascular endothelial cells with FOXO3a over expression. This study identified 419 DEGs between FOXO3a-OE HUVEC model and control cells. KEGG analysis indicated that the upregulated genes were mainly enriched in inflammation-related signaling pathways, and the downregulated genes were enriched in lipid metabolism-related pathways.
Collapse
Affiliation(s)
- Shajidan Abudureyimu
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Chunhui He
- China Heart Failure Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, 100010 Beijing, China
| | - Wei Xie
- Department of Cardiology, Xinjiang Production and Construction Corps Hospital, 830011 Urumqi, Xinjiang, China
| | - Zhuo Chen
- The Second Clinical Medical College of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Halisha Airikenjiang
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Dilihumaer Abulaiti
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Yan Cao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Cancer Hospital Xinjiang Medical University, 830000 Urumqi, Xinjiang, China
| | - Haitang Qiu
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Ying Gao
- Department of Comprehensive Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China.
| |
Collapse
|
2
|
Yang F, Zheng Y, Luo Q, Zhang S, Yang S, Chen X. Knockdown of NCAPD3 inhibits the tumorigenesis of non-small cell lung cancer by regulation of the PI3K/Akt pathway. BMC Cancer 2024; 24:408. [PMID: 38566039 PMCID: PMC10986035 DOI: 10.1186/s12885-024-12131-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] [Received: 07/03/2023] [Accepted: 03/16/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Accumulating evidence indicates that aberrant non-SMC condensin II complex subunit D3 (NCAPD3) is associated with carcinogenesis of various cancers. Nevertheless, the biological role of NCAPD3 in the pathogenesis of non-small cell lung cancer (NSCLC) remains unclear. METHODS Immunohistochemistry and Western blot were performed to assess NCAPD3 expression in NSCLC tissues and cell lines. The ability of cell proliferation, invasion, and migration was evaluated by CCK-8 assays, EdU assays, Transwell assays, and scratch wound healing assays. Flow cytometry was performed to verify the cell cycle and apoptosis. RNA-sequence and rescue experiment were performed to reveal the underlying mechanisms. RESULTS The results showed that the expression of NCAPD3 was significantly elevated in NSCLC tissues. High NCAPD3 expression in NSCLC patients was substantially associated with a worse prognosis. Functionally, knockdown of NCAPD3 resulted in cell apoptosis and cell cycle arrest in NSCLC cells as well as a significant inhibition of proliferation, invasion, and migration. Furthermore, RNA-sequencing analysis suggested that NCAPD3 contributes to NSCLC carcinogenesis by regulating PI3K/Akt/FOXO4 pathway. Insulin-like growth factors-1 (IGF-1), an activator of PI3K/Akt signaling pathway, could reverse NCAPD3 silence-mediated proliferation inhibition and apoptosis in NSCLC cells. CONCLUSION NCAPD3 suppresses apoptosis and promotes cell proliferation via the PI3K/Akt/FOXO4 signaling pathway, suggesting a potential use for NCAPD3 inhibitors as NSCLC therapeutics.
Collapse
Affiliation(s)
- Fan Yang
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, NO.29 of Xinquan Road, Gulou District, 350000, Fuzhou City, Fujian Province, China
| | - Yunfeng Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, NO.29 of Xinquan Road, Gulou District, 350000, Fuzhou City, Fujian Province, China
| | - Qiong Luo
- Department of Oncology, Fujian Medical University Union Hospital, NO.29 of Xinquan Road, Gulou District, 350000, Fuzhou City, Fujian Province, China
| | - Suyun Zhang
- Department of Internal Medicine, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China.
| | - Sheng Yang
- Department of Oncology, Fujian Medical University Union Hospital, NO.29 of Xinquan Road, Gulou District, 350000, Fuzhou City, Fujian Province, China.
| | - Xiangqi Chen
- Department of Respiratory Medicine, Fujian Medical University Union Hospital, NO.29 of Xinquan Road, Gulou District, 350000, Fuzhou City, Fujian Province, China.
| |
Collapse
|
3
|
Liou YJ, Chen MH, Hsu JW, Huang KL, Huang PH, Bai YM. Levels of circulating endothelial progenitor cells inversely correlate with manic and positive symptom severity in patients with bipolar disorder. Brain Behav 2022; 12:e2570. [PMID: 35481989 PMCID: PMC9226848 DOI: 10.1002/brb3.2570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/02/2022] [Accepted: 03/19/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Patients with bipolar disorder (BPD) are at high risk of cardiovascular diseases (CVDs) that are attributed to endothelial dysfunction. Circulating endothelial progenitor cells (cEPCs) are proposed as indicators of endothelial dysfunction. This study examined the relationship of cEPC numbers with BPD diagnosis and its clinical symptoms in patients with BPD. METHODS We recruited 48 patients with BPD and 50 healthy controls (HCs). All the patients had scores of <13 on the Young Mania Rating Scale (YMRS). In addition to the YMRS and Clinical Global Impression for BPD (CGI-BP), bipolar patients were assessed using relevant measurements for their depression, anxiety, general psychopathology, cognitive dysfunction and deficit, somatic symptoms, quality of life, and level of disability. cEPC counts were measured using flow cytometry. RESULTS The numbers of immature and mature cEPCs in the bipolar patients did not significantly differ from those in the HCs. After correction for multiple comparisons and controlling for body mass index and smokers, the number of immature cEPCs was observed to be inversely correlated with CGI-BP (corrected p [pcorr ] = .00018) and positive scores in the positive and negative syndrome scale (PANSS-P; pcorr = .0049). The number of mature cEPCs was inversely correlated with YMRS (pcorr = .014), CGI-BP (pcorr = .00022), and PANSS-P (pcorr = .0049) scores. In multivariate stepwise analysis, numbers of both types of cEPCs were associated with CGI-BP. CONCLUSIONS cEPC levels, an indicator of endothelial dysfunction and risk of CVDs, may be associated manic and positive symptom severities in patients with BPD.
Collapse
Affiliation(s)
- Ying-Jay Liou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ju-Wei Hsu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kai-Lin Huang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
4
|
Salvianolic Acid B Suppresses ER Stress-Induced NLRP3 Inflammasome and Pyroptosis via the AMPK/FoxO4 and Syndecan-4/Rac1 Signaling Pathways in Human Endothelial Progenitor Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8332825. [PMID: 35340217 PMCID: PMC8947883 DOI: 10.1155/2022/8332825] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 02/18/2022] [Indexed: 12/18/2022]
Abstract
Mounting evidence demonstrates uncontrolled endoplasmic reticulum (ER) stress responses can activate the inflammasome, which generally results in endothelial dysfunction, a major pathogenetic factor of chronic inflammatory diseases such as atherosclerosis. Salvianolic acid B (SalB), produced by Radix Salviae, exerts antioxidative and anti-inflammatory activities in multiple cell types. However, SalB's effects on ER stress-related inflammasome and endothelial dysfunction remain unknown. Here, we showed SalB substantially abrogated ER stress-induced cell death and reduction in capillary tube formation, with declined intracellular reactive oxygen species (ROS) amounts and restored mitochondrial membrane potential (MMP), as well as increased expression of HO-1 and SOD2 in bone marrow-derived endothelial progenitor cells (BM-EPCs). ER stress suppression by CHOP or caspase-4 siRNA transfection attenuated the protective effect of SalB. Additionally, SalB alleviated ER stress-mediated pyroptotic cell death via the suppression of TXNIP/NLRP3 inflammasome, as evidenced by reduced cleavage of caspase-1 and interleukin- (IL-) 1β and IL-18 secretion levels. Furthermore, this study provided a mechanistic basis that AMPK/FoxO4/KLF2 and Syndecan-4/Rac1/ATF2 signaling pathway modulation by SalB substantially prevented BM-EPCs damage associated with ER stress by decreasing intracellular ROS amounts and inducing NLRP3-dependent pyroptosis. In summary, our findings identify that ER stress triggered mitochondrial ROS release and NLRP3 generation in BM-EPCs, while SalB inhibits NLRP3 inflammasome-mediated pyroptotic cell death by regulating the AMPK/FoxO4/KLF2 and Syndecan-4/Rac1/ATF2 pathways. The current findings reveal SalB as a potential new candidate for the treatment of atherosclerotic heart disease.
Collapse
|
5
|
Qian J, Shen Q, Yan CX, Yin H, Cao X, Lin ZH, Cai YF, Liu H. Atorvastatin improves bone marrow endothelial progenitor cell function from patients with immune-related hemocytopenia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1142. [PMID: 34430583 PMCID: PMC8350688 DOI: 10.21037/atm-21-2459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/13/2021] [Indexed: 11/06/2022]
Abstract
Background Immune-related hemocytopenia (IRH) is a type of autoimmune disease that targets bone marrow hematopoietic cells. This study investigated the influence of atorvastatin on the functional aspects of bone marrow endothelial progenitor cells (BM EPCs) in IRH patients. Methods BM EPCs were isolated from 15 patients with IRH and 20 normal controls. The isolated BM EPCs were characterized by flow cytometry. Cell counting kit-8, flow cytometry, and Transwell migration assays were used to determine the proliferation, apoptosis, and migration of BM EPCs, respectively. Protein levels were determined by western blot assay. Results The BM EPCs isolated from IRH patients showed reduced proliferation, increased apoptosis, and attenuated migratory ability compared to those from normal controls. Western blot analysis showed that the protein level of p-p38 was significantly increased, while that of Phosphorylated protein kinase B (p-AKT) was significantly decreased in the BM EPCs from IRH patients, compared to BM EPCs from healthy subjects. Cell proliferation and migration were significantly enhanced by atorvastatin, recombinant human thrombopoietin, and SB20358 compared to the untreated BM EPCs from IRH patients. Atorvastatin, Recombinant human thrombopoietin (TPO), and SB20358 treatment significantly suppressed the protein levels of p-p38 protein, but increased those of p-AKT in BM EPCS from IRH patients. Conclusions In summary, atorvastatin increases the number and function of BM EPCs in IRH patients by regulating the p38 and AKT signaling pathways.
Collapse
Affiliation(s)
- Juan Qian
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qian Shen
- Department of Oncology, Nantong Oncology Hospital, Nantong, China
| | | | - Hong Yin
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Cao
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zeng-Hua Lin
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yi-Feng Cai
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
6
|
Wang X, Li X, Wang Z. lncRNA MEG3 inhibits pituitary tumor development by participating in cell proliferation, apoptosis and EMT processes. Oncol Rep 2021; 45:40. [PMID: 33649837 PMCID: PMC7934213 DOI: 10.3892/or.2021.7991] [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: 06/22/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
Pituitary tumors do not pose a threat to life but can cause visual disturbances and serious clinical syndromes, such as infertility and metabolic syndrome. Therefore, screening of key genes involved in the occurrence and development of pituitary tumors can provide new targets for the treatment of pituitary tumors. The aim of the present study was to investigate the molecular mechanism of long non‑coding (lnc.) RNA maternally expressed 3 (MEG3) in cell proliferation, apoptosis and epithelial‑mesenchymal transition (EMT) processes of pituitary tumor. Tissue samples were obtained from 34 patients who underwent surgical treatment of pituitary tumors. Pituitary tumor cells (GH3 and MMQ) were transfected with pcDNA3.1(+)‑MEG3, short hairpin (sh)MEG3, microRNA (miR)‑23‑3p inhibitor or their controls using Lipofectamine® 2000. Reverse transcription‑quantitative PCR and western blot analyses were used to detect the levels of MEG3, miR‑23b‑3p and FOXO4, as well as proliferation‑, apoptosis‑ and EMT‑associated genes and proteins. Cell Counting Kit‑8 and flow cytometry assays were performed to detect proliferation and apoptosis, and Transwell assay was undertaken to assess invasion and migration. Luciferase reporter and RNA pulldown assays were performed to verify the binding between lncRNA MEG3, miR‑23b‑3p and FOXO4. Pearson's correlation analysis was used to analyze the correlation between expression levels of MEG3, miR‑23b‑3p and FOXO4. lncRNA MEG3 was expressed at lower levels in pituitary tumor tissues and cells. Overexpression of lncRNA MEG3 inhibited proliferation, invasion and migration and accelerated apoptosis of pituitary tumor cells. lncRNA MEG3 negatively regulated miR‑23b‑3p expression levels, while miR‑23b‑3p negatively regulated FOXO4 expression levels. Overexpression of lncRNA MEG3 inhibited the EMT process in pituitary tumor cells. miR‑23‑3p inhibitor rescued the effect of shMEG3 on proliferation, invasion, migration, apoptosis and the EMT process in pituitary tumor cells. lncRNA MEG3 inhibited pituitary tumor development by participating in cell proliferation, apoptosis and the EMT process, which may present a novel target for pituitary tumor treatment.
Collapse
Affiliation(s)
- Xuejian Wang
- Department of Neurosurgery, First Affiliated Hospital of Soochow University, Soochow University, Soochow, Jiangsu 225000, P.R. China
| | - Xiangdong Li
- Department of Neurosurgery, First Affiliated Hospital of Soochow University, Soochow University, Soochow, Jiangsu 225000, P.R. China
| | - Zhifeng Wang
- Department of Neurosurgery, Affiliated Hospital 2 of Nantong University, Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
7
|
Wu Q, Qi B, Duan X, Ming X, Yan F, He Y, Bu X, Sun S, Zhu H. MicroRNA-126 enhances the biological function of endothelial progenitor cells under oxidative stress via PI3K/Akt/GSK3β and ERK1/2 signaling pathways. Bosn J Basic Med Sci 2021; 21:71-80. [PMID: 31999938 PMCID: PMC7861621 DOI: 10.17305/bjbms.2019.4493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 12/26/2019] [Indexed: 12/15/2022] Open
Abstract
Endothelial progenitor cell (EPC) transplantation is a safe and effective method to treat acute myocardial infarction (AMI). However, oxidative stress leads to the death of a large number of EPCs in the early stage of transplantation, severely weakening the therapeutic effect. Previous studies demonstrated that microRNAs regulate the biological function of EPCs. The aim of the current study was to investigate the effect of microRNA on the biological function of EPCs under oxidative stress. Quantitative reverse transcription PCR was performed to detect the expression of miR-126, miR-508-5p, miR-150, and miR-16 in EPCs from rats, among which miR-126 showed a relatively higher expression. Treatment with H2O2 decreased miR-126 expression in EPCs in a dose-dependent manner. EPCs were further transfected with miR-126 mimics or inhibitors, followed by H2O2 treatment. Overexpression of miR-126 enhanced the proliferation, migration, and tube formation of H2O2-treated EPCs. MiR-126 overexpression also inhibited reactive oxygen species and malondialdehyde levels and enhanced superoxide dismutase levels, as well as increased angiopoietin (Ang)1 expression and decreased Ang2 expression in H2O2-treated EPCs. Moreover, miR-126 participated in the regulation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in EPCs, where both pathways were activated after miR-126 overexpression in H2O2-treated EPCs. Overall, we showed that miR-126 promoted the biological function of EPCs under H2O2-induced oxidative stress by activating the PI3K/Akt/GSK3β and ERK1/2 signaling pathway, which may serve as a new therapeutic approach to treat AMI.
Collapse
Affiliation(s)
- Qinqin Wu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Benling Qi
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Duan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Ming
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengqin Yan
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxia He
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofen Bu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Sun
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Gerontology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
L. Michmerhuizen N, Wang J, Brenner J. Integrated Molecular Profiling as an Approach to Identify PI3K Inhibitor Resistance Mechanisms. Mol Pharmacol 2020. [DOI: 10.5772/intechopen.92875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The identification of drug resistance pathways and approaches to target these pathways remains a significant and important challenge in cancer biology. Here, we address this challenge in the context of ongoing efforts to advance phosphatidylinositol 3-kinase (PI3K) inhibitors for the treatment of PI3K-aberrant cancers. While PI3K inhibitors have had tremendous success in some diseases, such as breast cancer, early clinical trials in other malignancies, such as head and neck squamous cell carcinoma (HNSCC), have not had the same level of success. Since HNSCC and other cancers display relatively high PI3K pathway alteration rates (>45%), these underwhelming results suggest that additional or unexpected factors may contribute to the lower response rates. Here, we highlight some of the emerging functional genomic and sequencing approaches being used to identify predictive biomarkers of PI3K inhibitor response using both cancer cell lines and clinical trial specimens. Importantly, these approaches have uncovered both innate genetic and adaptive mechanisms driving PI3K inhibitor resistance. In this chapter, we describe recent technological advances that have revolutionized our understanding of PI3K inhibitor resistance pathways in HNSCC and highlight how these and other approaches lay the groundwork to make significant strides in our understanding of molecular pharmacology in the cancer field.
Collapse
|
9
|
Li X, Chen X, Liu Y, Zhang P, Zheng Y, Zeng W. The Histone Methyltransferase SETDB1 Modulates Survival of Spermatogonial Stem/Progenitor Cells Through NADPH Oxidase. Front Genet 2020; 11:997. [PMID: 33133132 PMCID: PMC7567028 DOI: 10.3389/fgene.2020.00997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022] Open
Abstract
SETDB1, a histone H3 lysine 9 (H3K9) methyltransferase, is crucial in meiosis and embryo development. This study aimed to investigate whether SETDB1 was associated with spermatogonial stem cells (SSC) homeostasis. We found that knockdown of Setdb1 impaired cell proliferation, led to an increase in reactive oxygen species (ROS) level through NADPH oxidase, and Setdb1 deficiency activated ROS downstream signaling pathways, including JNK and p38 MAPK, which possibly contributed to SSC apoptosis. Melatonin scavenged ROS and rescued the phenotype of Setdb1 KD. In addition, we demonstrated that SETDB1 regulated NADPH oxidase 4 (Nox4) and E2F1. Therefore, this study uncovers the new roles of SETDB1 in mediating intracellular ROS homeostasis for the survival of SSC.
Collapse
Affiliation(s)
- Xueliang Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaoxu Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yingdong Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Pengfei Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yi Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wenxian Zeng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
10
|
Farhan M, Silva M, Xingan X, Huang Y, Zheng W. Role of FOXO Transcription Factors in Cancer Metabolism and Angiogenesis. Cells 2020; 9:E1586. [PMID: 32629884 PMCID: PMC7407656 DOI: 10.3390/cells9071586] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
Forkhead box O transcription factors (FOXOs) regulate several signaling pathways and play crucial roles in health and disease. FOXOs are key regulators of the expression of genes involved in multiple cellular processes and their deregulation has been implicated in cancer. FOXOs are generally considered tumor suppressors and evidence also suggests that they may have a role in the regulation of cancer metabolism and angiogenesis. In order to continue growing and proliferating, tumor cells have to reprogram their metabolism and induce angiogenesis. Angiogenesis refers to the process of new blood capillary formation from pre-existing vessels, which is an essential driving force in cancer progression and metastasis through supplying tumor cells with oxygen and nutrients. This review summarizes the roles of FOXOs in the regulation of cancer metabolism and angiogenesis. A deeper knowledge of the involvement of FOXOs in these two key processes involved in cancer dissemination may help to develop novel therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Mohd Farhan
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China; (M.F.); (M.S.); (X.X.)
| | - Marta Silva
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China; (M.F.); (M.S.); (X.X.)
| | - Xing Xingan
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China; (M.F.); (M.S.); (X.X.)
| | - Yu Huang
- Heart and Vascular Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China;
| | - Wenhua Zheng
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China; (M.F.); (M.S.); (X.X.)
| |
Collapse
|
11
|
Emmi G, Mannucci A, Argento FR, Silvestri E, Vaglio A, Bettiol A, Fanelli A, Stefani L, Taddei N, Prisco D, Fiorillo C, Becatti M. Stem-Cell-Derived Circulating Progenitors Dysfunction in Behçet's Syndrome Patients Correlates With Oxidative Stress. Front Immunol 2019; 10:2877. [PMID: 31921141 PMCID: PMC6923242 DOI: 10.3389/fimmu.2019.02877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 11/22/2019] [Indexed: 12/29/2022] Open
Abstract
Behçet's syndrome (BS) is a systemic vasculitis considered as the prototype of a systemic inflammation-induced thrombotic condition whose pathogenesis cannot be explained just by coagulation abnormalities. Circulating hematopoietic progenitor cells (CPC), a population of rare, pre-differentiated adult stem cells originating in the bone marrow and capable of both self-renewal and multi-lineage differentiation, are mobilized in response to vascular injury and play a key role in tissue repair. In cardiovascular and thrombotic diseases, low circulating CPC number and reduced CPC function have been observed. Oxidative stress may be one of the relevant culprits that account for the dysfunctional and numerically reduced CPC in these conditions. However, the detailed mechanisms underlying CPC number reduction are unknown. On this background, the present study was designed to evaluate for the first time the possible relationship between CPC dysfunction and oxidative stress in BS patients. In BS patients, we found signs of plasma oxidative stress and significantly lower CD34+/CD45−/dim and CD34+/CD45−/dim/CD133+ CPC levels. Importantly, in all the considered CPC subsets, significantly higher ROS levels with respect to controls were observed. Higher levels of caspase-3 activity in all the considered CPC population and a strong reduction in GSH content in CPC subpopulation from BS patients with respect to controls were also observed. Interestingly, in BS patients, ROS significantly correlated with CPC number and CPC caspase-3 activity and CPC GSH content significantly correlated with CPC number, in all CPC subsets. Collectively, these data demonstrate for the first time that CPC from BS patients show signs of oxidative stress and apoptosis and that a reduced CPC number is present in BS patients with respect to controls. Interestingly, we observed an inverse correlation between circulating CPC number and CPC ROS production, suggesting a possible toxic ROS effect on CPC in BS patients. The significant correlations between ROS production/GSH content and caspase-3 activity point out that oxidative stress can represent a determinant in the onset of apoptosis in CPC. These data support the hypothesis that oxidative-stress-mediated CPC dysfunctioning may counteract their vascular repair actions, thereby contributing to the pathogenesis and the progression of vascular disease in BS.
Collapse
Affiliation(s)
- Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Amanda Mannucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Flavia Rita Argento
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Augusto Vaglio
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Alessandra Bettiol
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Alessandra Fanelli
- Central Laboratory, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Laura Stefani
- Department of Clinical and Experimental Medicine, Center of Sports Medicine, University of Firenze, Firenze, Italy
| | - Niccolò Taddei
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Firenze, Firenze, Italy
| |
Collapse
|
12
|
Xue M, Joo YA, Li S, Niu C, Chen G, Yi X, Liang Y, Chen Z, Shen Y, Ye W, Cai L, Wang X, Jin L, Cong W. Metallothionein Protects the Heart Against Myocardial Infarction via the mTORC2/FoxO3a/Bim Pathway. Antioxid Redox Signal 2019; 31:403-419. [PMID: 30860395 DOI: 10.1089/ars.2018.7597] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aims: Cardiac-specific overexpression of metallothionein (MT) has been shown to be beneficial in ischemic heart disease, but the detailed mechanisms through which MT protects against myocardial infarction (MI) remain unknown. This study assessed the involvement of the mTORC2/FoxO3a/Bim pathway in the cardioprotective effects of MT. Results: MI was induced in wild-type (FVB) mice and in cardiac-specific MT-overexpressing transgenic (MT-TG) mice by ligation of the left anterior descending (LAD) coronary artery. Cardiac function was better; infarct size and cardiomyocyte apoptosis were lower in MT-TG mice than in FVB mice after MI. Moreover, MT-TG mice exhibited better phenotypes after LAD ligation than FVB mice treated with Mn(III)tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride (MnTMPyP; a reactive oxygen species [ROS] scavenger) and cardiac-specific catalase-overexpressing transgenic (CAT-TG) mice, which showed the same ROS levels as MT-TG mice after MI. Activation of mechanistic target of rapamycin complex 2 (mTORC2) was essential for the cardioprotective effects of MT against MI. In addition, MT attenuated the downregulation of phospho-FoxO3a after MI, inhibiting the expression of the apoptosis-associated gene Bim, located downstream of FoxO3a, and reducing the level of apoptosis after MI. To mimic ischemic-injured FVB and MT-TG mice in vitro, H9c2 and MT-overexpressing H9c2 (H9c2MT7) cardiomyocytes were subjected to oxygen and glucose deprivation, with the results being consistent with those obtained in vivo. Innovation and Conclusion: The cardioprotective effects of MT against MI are not entirely dependent upon its ability to eliminate ROS. Rather, MT overexpression mostly protects against MI through the mTORC2-FoxO3a-Bim pathway.
Collapse
Affiliation(s)
- Mei Xue
- 1 Precision Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Young A Joo
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Santie Li
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chao Niu
- 3 The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Gen Chen
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xinchu Yi
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yangzhi Liang
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhiwei Chen
- 3 The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yingjie Shen
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Weijian Ye
- 3 The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Lu Cai
- 4 Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville, Louisville, Kentucky
| | - Xu Wang
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Litai Jin
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Weitao Cong
- 2 School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
13
|
Xu A, Chen J, Peng H, Han G, Cai H. Simultaneous Interrogation of Cancer Omics to Identify Subtypes With Significant Clinical Differences. Front Genet 2019; 10:236. [PMID: 30984238 PMCID: PMC6448130 DOI: 10.3389/fgene.2019.00236] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/04/2019] [Indexed: 11/21/2022] Open
Abstract
Recent advances in high-throughput sequencing have accelerated the accumulation of omics data on the same tumor tissue from multiple sources. Intensive study of multi-omics integration on tumor samples can stimulate progress in precision medicine and is promising in detecting potential biomarkers. However, current methods are restricted owing to highly unbalanced dimensions of omics data or difficulty in assigning weights between different data sources. Therefore, the appropriate approximation and constraints of integrated targets remain a major challenge. In this paper, we proposed an omics data integration method, named high-order path elucidated similarity (HOPES). HOPES fuses the similarities derived from various omics data sources to solve the dimensional discrepancy, and progressively elucidate the similarities from each type of omics data into an integrated similarity with various high-order connected paths. Through a series of incremental constraints for commonality, HOPES can take both specificity of single data and consistency between different data types into consideration. The fused similarity matrix gives global insight into patients' correlation and efficiently distinguishes subgroups. We tested the performance of HOPES on both a simulated dataset and several empirical tumor datasets. The test datasets contain three omics types including gene expression, DNA methylation, and microRNA data for five different TCGA cancer projects. Our method was shown to achieve superior accuracy and high robustness compared with several benchmark methods on simulated data. Further experiments on five cancer datasets demonstrated that HOPES achieved superior performances in cancer classification. The stratified subgroups were shown to have statistically significant differences in survival. We further located and identified the key genes, methylation sites, and microRNAs within each subgroup. They were shown to achieve high potential prognostic value and were enriched in many cancer-related biological processes or pathways.
Collapse
Affiliation(s)
- Aodan Xu
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - Jiazhou Chen
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - Hong Peng
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - GuoQiang Han
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - Hongmin Cai
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
14
|
Chou RH, Chen CY, Chen IC, Huang HL, Lu YW, Kuo CS, Chang CC, Huang PH, Chen JW, Lin SJ. Trimethylamine N-Oxide, Circulating Endothelial Progenitor Cells, and Endothelial Function in Patients with Stable Angina. Sci Rep 2019; 9:4249. [PMID: 30862856 PMCID: PMC6414518 DOI: 10.1038/s41598-019-40638-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Trimethylamine N-oxide (TMAO) is a metabolite originated from bacterial metabolism of choline-rich foods. Evidence suggests an association between TMAO and atherosclerosis, but the relationship between TMAO and endothelial progenitor cells (EPCs) remains unclear. This study aimed to identify the relationship between TMAO concentrations, circulating EPCs, and endothelial function in patients with stable angina. Eighty-one stable angina subjects who underwent coronary angiography were enrolled. The circulating EPCs and flow-mediated vasodilation (FMD) were measured to evaluate endothelial function. Plasma TMAO and inflammatory markers, such as hsCRP and IL-1β, were determined. Furthermore, the effect of TMAO on EPCs was assessed in vitro. Patients with lower FMD had significantly decreased circulating EPCs, elevated TMAO, hsCRP, and IL-1β concentrations. Plasma TMAO levels were negatively correlated with circulating EPC numbers and the FMD, and positively correlated with hsCRP, IL-1β concentrations. In in vitro studies, incubation of TMAO in cultured EPCs promoted cellular inflammation, elevated oxidative stress, and suppressed EPC functions. Enhanced plasma TMAO levels were associated with reduced circulating EPCs numbers, endothelial dysfunction, and more adverse cardiovascular events. These findings provided evidence of TMAO’s toxicity on EPCs, and delivered new insight into the mechanism of TMAO-mediated atherosclerosis, which could be derived from TMAO-downregulated EPC functions.
Collapse
Affiliation(s)
- Ruey-Hsing Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Yu Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - I-Chun Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Lei Huang
- Institute of Physiology, National Yang-Ming University, Taipei, Taiwan.,Department of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Ya-Wen Lu
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Sung Kuo
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan. .,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Chun-Chin Chang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan. .,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jaw-Wen Chen
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute and Department of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Xie M, Yang A, Ma J, Wu M, Xu H, Wu K, Jin Y, Xie Y. Akt2 mediates glucocorticoid resistance in lymphoid malignancies through FoxO3a/Bim axis and serves as a direct target for resistance reversal. Cell Death Dis 2019; 9:1013. [PMID: 30598523 PMCID: PMC6312545 DOI: 10.1038/s41419-018-1043-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 01/22/2023]
Abstract
Glucocorticoids (GCs) are widely used drugs in the treatment of lymphoid malignancies; resistance of GCs in lymphocytes confers poor prognosis and the mechanisms are poorly understood. Here, we found T-acute lymphoblastic leukemia (T-ALL) cells acquire resistance to dexamethasone (DEX)-mediated killing through abnormal activation of Akt, resulting in inhibition of the FoxO3a/Bim pathway. The resistant state was reported to be associated with increased glycolysis, NOTCH1 activating mutations and activated PI3K/ serum GS regulated kinases (SGK) pathway. Use of aforementioned pathway inhibitors blocked FoxO3a-phosphorylation and partially improved DEX-mediated killing of GC-resistant T-ALL cells, further revealing the essential role of the FoxO3a/Bim pathway in the development of GC resistance. Inhibition of Akt is most effective at restoring sensitivity to DEX of GC-resistant lymphocytes in vitro and in vivo, but shows significant hepatotoxicity in vivo. A significantly elevated expression of Akt2 not Akt1 in intrinsically, secondarily GC-resistant lymphocytes and relapsed/refractory ALL patients implicates a more specific target for GC resistance. Mechanistically, Akt2 has a stronger binding capacity with FoxO3a compared to Akt1, and acts as a direct and major negative regulator of FoxO3a activity driving GC resistance. Pharmacologic inhibition of Akt2 more effectively restores sensitivity to GCs than inhibition of Akt1 in vitro, shows higher synergistic effect acting with DEX, and reverses GC resistance in GC-resistant T- or B- lymphoid tumors in vivo with reduced liver toxicity. In summary, these results suggest that Akt2 might serve as a more direct and specific kinase mediating GC resistance through FoxO3a/Bim signaling pathway, and Akt2 inhibition may be explored as a promising target for treating GC-resistant hematopoietic malignancies.
Collapse
Affiliation(s)
- Mixue Xie
- Senior Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Apeng Yang
- Department of Hematology & Rheumatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Jiexian Ma
- Department of Hematology & Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Min Wu
- Department of Hematology & Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Hongyue Xu
- Department of Hematology & Oncology, The Second People's Hospital of Liaocheng, Liaocheng, 252600, China
| | - Kefei Wu
- Department of Hematology & Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Youxin Jin
- The School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Yanhui Xie
- Department of Hematology & Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| |
Collapse
|
16
|
FOXO3a-dependent up-regulation of Mxi1-0 promotes hypoxia-induced apoptosis in endothelial cells. Cell Signal 2018; 51:233-242. [DOI: 10.1016/j.cellsig.2018.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
|
17
|
Liu ZQ, Du JJ, Ren JJ, Zhang ZY, Guo XB, Yan YE, Jia XT, Gu NB, Di ZL, Li SZ. miR-183-96-182 clusters alleviated ox-LDL-induced vascular endothelial cell apoptosis in vitro by targeting FOXO1. RSC Adv 2018; 8:35031-35041. [PMID: 35547044 PMCID: PMC9087689 DOI: 10.1039/c8ra06866f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the role of FOXO1 and miR-183-96-182 clusters in ox-LDL induced endothelial cell apoptosis. METHODS FOXO1 overexpression (OE) and knockdown (KD) as well as AKT1 OE in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs) were achieved by lentiviral transduction. Upregulation of miR-183-5p, miR-182-5p or miR-96-5p was mimicked by agomir treatment. FOXO1 gene transcription was monitored by FOXO1 promotor reporter assay. Cell apoptosis in culture was monitored by TiterTACS in situ detection. Regulation of FOXO1 gene expression by an miRNA targeting mechanism was monitored by AGO2-RNA immunoprecipitation assay. RESULTS FOXO1 mRNA and protein expression levels in ox-LDL treated HUVECs or HAECs were significantly upregulated due to transcriptional and miRNA targeting mechanisms. MiR-183-5p, miR-182-5p and miR-96-5p expression levels in HUVECs or HAECs were significantly reduced by ox-LDL treatment, the overexpression of which by agomir treatment partially reduced the FOXO1 mRNA/protein expression levels and cell apoptosis which was upregulated by ox-LDL treatment. FOXO1 overexpression antagonized the effect of the agomir treatment indicated above. MiR-183-5p, miR-182-5p and miR-96-5p agomir treatment partially rescued the FOXO1 pSer256/total FOXO1 protein ratio and the AKT1 pSer473 level that were reduced by ox-LDL treatment in the HUVECs or HAECs. AKT1 overexpression significantly reduced FOXO1 protein expression, increased miR-182-5p and miR-183-5p expression, and partially alleviated ox-LDL induced HUVEC or HAEC apoptosis in an miR-183-5p and miR-182-5p-dependent manner. CONCLUSION miR-183-96-182 clusters could partially alleviate ox-LDL-induced apoptosis in HUVECs or HAECs by targeting FOXO1.
Collapse
Affiliation(s)
- Zhi-Qin Liu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Jing-Jing Du
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Jing-Jing Ren
- Department of Hematology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Zhi-Yong Zhang
- Department of Neurology, China-Japan Friendship Hospital Beijing 100029 China
| | - Xiao-Bo Guo
- Department of Hematology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Yu-E Yan
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Xiao-Tao Jia
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Nai-Bing Gu
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - Zheng-Li Di
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University, School of Medicine Xi'an 710003 Shaanxi China
| | - San-Zhong Li
- Department of Neurosurgery, Xi-jing Hospital Fourth Military Medical University No. 127, Changle Xi Road Xi'an 710032 Shaanxi China +86-185-9140-9510
| |
Collapse
|
18
|
Pan J, Di YQ, Li YB, Chen CH, Wang JX, Zhao XF. Insulin and 20-hydroxyecdysone oppose each other in the regulation of phosphoinositide-dependent kinase-1 expression during insect pupation. J Biol Chem 2018; 293:18613-18623. [PMID: 30305395 DOI: 10.1074/jbc.ra118.004891] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/26/2018] [Indexed: 12/17/2022] Open
Abstract
Insulin promotes larval growth of insects by stimulating the synthesis of the steroid hormone 20-hydroxyecdysone (20E), which induces pupation and apoptosis. However, the mechanism underlying the coordinate regulation of insect pupation and apoptosis by these two functionally opposing hormones is still unclear. Here, using the lepidopteran insect and serious agricultural pest Helicoverpa armigera (cotton bollworm) as a model, we report that phosphoinositide-dependent kinase-1 (PDK1) and forkhead box O (FoxO) play key roles in these processes. We found that the transcript levels of the PDK1 gene are increased during the larval feeding stages. Moreover, PDK1 expression was increased by insulin, but repressed by 20E. dsRNA-mediated PDK1 knockdown in the H. armigera larvae delayed pupation and resulted in small pupae and also decreased Akt/protein kinase B expression and increased FoxO expression. Furthermore, the PDK1 knockdown blocked midgut remodeling and decreased 20E levels in the larvae. Of note, injecting larvae with 20E overcame the effect of the PDK1 knockdown and restored midgut remodeling. FoxO overexpression in an H. armigera epidermal cell line (HaEpi) did not induce apoptosis, but promoted autophagy and repressed cell proliferation. These results reveal cross-talk between insulin and 20E and that both hormones oppose each other's activities in the regulation of insect pupation and apoptosis by controlling PDK1 expression and, in turn, FoxO expression. We conclude that sufficiently high 20E levels are a key factor for inducing apoptosis during insect pupation.
Collapse
Affiliation(s)
- Jing Pan
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Yu-Qin Di
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Yong-Bo Li
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Cai-Hua Chen
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Jin-Xing Wang
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Xiao-Fan Zhao
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
19
|
Jiang S, Yang Z, Di S, Hu W, Ma Z, Chen F, Yang Y. Novel role of forkhead box O 4 transcription factor in cancer: Bringing out the good or the bad. Semin Cancer Biol 2018; 50:1-12. [DOI: 10.1016/j.semcancer.2018.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 04/28/2018] [Indexed: 10/17/2022]
|
20
|
Atorvastatin enhances bone marrow endothelial cell function in corticosteroid-resistant immune thrombocytopenia patients. Blood 2018; 131:1219-1233. [PMID: 29288170 DOI: 10.1182/blood-2017-09-807248] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/18/2017] [Indexed: 12/15/2022] Open
Abstract
Key Points
Impaired BM EPCs were found in corticosteroid-resistant ITP patients. Atorvastatin improved BM EPC quantity and function, representing a novel therapy approach for corticosteroid-resistant ITP patients.
Collapse
|
21
|
Zhang XQ, Dong JJ, Cai T, Shen X, Zhou XJ, Liao L. High glucose induces apoptosis via upregulation of Bim expression in proximal tubule epithelial cells. Oncotarget 2018; 8:24119-24129. [PMID: 28445931 PMCID: PMC5421832 DOI: 10.18632/oncotarget.15491] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 02/02/2017] [Indexed: 02/04/2023] Open
Abstract
Diabetic nephropathy is the primary cause of end-stage renal disease. Apoptosis of tubule epithelial cells is a major feature of diabetic nephropathy. The mechanisms of high glucose (HG) induced apoptosis are not fully understood. Here we demonstrated that, HG induced apoptosis via upregulating the expression of proapoptotic Bcl-2 homology domain 3 (BH3)-only protein Bim protein, but not bring a significant change in the baseline level of autophagy in HK2 cells. The increase of Bim expression was caused by the ugregulation of transcription factors, FOXO1 and FOXO3a. Bim expression initiates BAX/BAK-mediated mitochondria-dependent apoptosis. Silence of Bim by siRNA in HK2 cells prevented HG-induced apoptosis and also sensitized HK2 cells to autophagy during HG treatment. The autophagy inhibitor 3-MA increased the injury in Bim knockdown HK2 cells by retriggering apoptosis. The above results suggest a Bim-independent apoptosis pathway in HK2 cells, which normally could be inhibited by autophagy. Overall, our results indicate that HG induces apoptosis via up-regulation of Bim expression in proximal tubule epithelial cells.
Collapse
Affiliation(s)
- Xiao-Qian Zhang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Jian-Jun Dong
- Department of Endocrinology, Qilu Hospital of Shandong University, Shandong, Jinan, China
| | - Tian Cai
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Medicine, Tai'an Hospital of Traditional Chinese Medicine, Tai'an, Shandong, China
| | - Xue Shen
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Jun Zhou
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
22
|
The balance between induction and inhibition of mevalonate pathway regulates cancer suppression by statins: A review of molecular mechanisms. Chem Biol Interact 2017; 273:273-285. [PMID: 28668359 DOI: 10.1016/j.cbi.2017.06.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/06/2017] [Accepted: 06/22/2017] [Indexed: 12/14/2022]
Abstract
Statins are widely used drugs for their role in decreasing cholesterol in hypercholesterolemic patients. Statins through inhibition of Hydroxy Methyl Glutaryl-CoA Reductase (HMGCR), the main enzyme of the cholesterol biosynthesis pathway, inhibit mevalonate pathway that provides isoprenoids for prenylation of different proteins such as Ras superfamily which has an essential role in cancer developing. Inhibition of the mevalonate/isoprenoid pathway is the cause of the cholesterol independent effects of statins or pleotropic effects. Depending on their penetrance into the extra-hepatic cells, statins have different effects on mevalonate/isoprenoid pathway. Lipophilic statins diffuse into all cells and hydrophilic ones use a variety of membrane transporters to gain access to cells other than hepatocytes. It has been suggested that the lower accessibility of statins for extra-hepatic tissues may result in the compensatory induction of mevalonate/isoprenoid pathway and so cancer developing. However, most of the population-based studies have demonstrated that statins have no effect on cancer developing, even decrease the risk of different types of cancer. In this review we focus on the cancer developing "potentials" and the anti-cancer "activities" of statins regarding the effects of statins on mevalonate/isoprenoid pathway in the liver and extra-hepatic tissues.
Collapse
|
23
|
Effect of Kangshuanyihao Formula on the Inflammatory Reaction and SIRT1/TLR4/NF- κB Signaling Pathway in Endothelial Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:9019765. [PMID: 28539968 PMCID: PMC5429935 DOI: 10.1155/2017/9019765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
Endothelial injury plays an important role in atherosclerosis (AS). Kangshuanyihao formula uses therapeutic principles from Chinese medicine to supplement Qi, thereby promoting blood circulation, and remove blood stasis. The mechanism by which the formula inhibits endothelial injury was examined in a rat model of 1,25-dihydroxyvitamin D3 (VD3) intraperitoneal injection and high-fat-induced endothelial injury. Rats were randomly divided into the model, high-dose, middle-dose, low-dose, positive drug (rosuvastatin), and combination (positive drug + middle-dose) groups; 10 Sprague-Dawley rats served as the blank group. The aortic endothelium was stained with hematoxylin and eosin and the levels of blood lipids and inflammation markers (mRNA and protein) were measured. Endothelial injury, lipid levels, and inflammation were increased in the model. Kangshuanyihao formula reduced endothelial injury, improved lipid levels, and downregulated inflammation, as shown by significant reduction of the protein levels of SIRT1, TLR4, and NF-κB and mRNA levels of SIRT1, TLR4, NF-κB, IL-1β, IL-6, and IL-12. Thus, we conclude that Kangshuanyihao formula can inhibit the inflammatory reaction in the rat model of high-fat-induced endothelial injury after intraperitoneal injection of VD3. This mechanism may be attributed to regulating the SIRT1/TLR4/NF-κB signaling pathway.
Collapse
|
24
|
Sun J, Ren D. IER3IP1 deficiency leads to increased β-cell death and decreased β-cell proliferation. Oncotarget 2017; 8:56768-56779. [PMID: 28915629 PMCID: PMC5593600 DOI: 10.18632/oncotarget.18179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Mutations in the gene for Immediate Early Response 3 Interacting Protein 1 (IER3IP1) cause permanent neonatal diabetes mellitus in human. The mechanisms involved have not been determined and the role of IER3IP1 in β-cell survival has not been characterized. In order to determine if there is a molecular link between IER3IP1 deficiency and β-cell survival and proliferation, we knocked down Ier3ip1 gene expression in mouse MIN6 insulinoma cells. IER3IP1 suppression induced apoptotic cell death which was associated with an increase in Bim and a decrease in Bcl-xL. Knockdown of Bim reduced apoptotic cell death in MIN6 cells induced by IER3IP1 suppression. Overexpression of the anti-apoptotic molecule Bcl-xL prevents cell death induced by IER3IP1 suppression. Moreover, IER3IP1 also regulates activation of the unfolded protein response (UPR). IER3IP1 suppression impairs the Inositol Requiring 1 (IRE1) and PKR-like ER kinase (PERK) arms of UPR. The cell proliferation of MIN6 cells was also decreased in IER3IP1 deficient cells. These results suggest that IER3IP1 suppression induces an increase in cell death and a decrease in cell proliferation in MIN6 cells, which may be the mechanism that mutations in IER3IP1 lead to diabetes.
Collapse
Affiliation(s)
- Juan Sun
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Decheng Ren
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
25
|
Mercado-Matos J, Clark JL, Piper AJ, Janusis J, Shaw LM. Differential involvement of the microtubule cytoskeleton in insulin receptor substrate 1 (IRS-1) and IRS-2 signaling to AKT determines the response to microtubule disruption in breast carcinoma cells. J Biol Chem 2017; 292:7806-7816. [PMID: 28320862 DOI: 10.1074/jbc.m117.785832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Indexed: 01/17/2023] Open
Abstract
The insulin receptor substrate (IRS) proteins serve as essential signaling intermediates for the activation of PI3K by both the insulin-like growth factor 1 receptor (IGF-1R) and its close family member, the insulin receptor (IR). Although IRS-1 and IRS-2 share significant homology, they regulate distinct cellular responses downstream of these receptors and play divergent roles in breast cancer. To investigate the mechanism by which signaling through IRS-1 and IRS-2 results in differential outcomes, we assessed the involvement of the microtubule cytoskeleton in IRS-dependent signaling. Treatment with drugs that either stabilize or disrupt microtubules reveal that an intact microtubule cytoskeleton contributes to IRS-2- but not IRS-1-mediated activation of AKT by IGF-1. Proximal IGF-1R signaling events, including IRS tyrosine phosphorylation and recruitment of PI3K, are not inhibited by microtubule disruption, indicating that IRS-2 requires the microtubule cytoskeleton at the level of downstream effector activation. IRS-2 colocalization with tubulin is enhanced upon Taxol-mediated microtubule stabilization, which, together with the signaling data, suggests that the microtubule cytoskeleton may facilitate access of IRS-2 to downstream effectors such as AKT. Of clinical relevance is that our data reveal that expression of IRS-2 sensitizes breast carcinoma cells to apoptosis in response to treatment with microtubule-disrupting drugs, identifying IRS-2 as a potential biomarker for the response of breast cancer patients to Vinca alkaloid drug treatment.
Collapse
Affiliation(s)
- Jose Mercado-Matos
- From the Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jennifer L Clark
- From the Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Andrew J Piper
- From the Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Jenny Janusis
- From the Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Leslie M Shaw
- From the Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
26
|
Sandhu K, Mamas M, Butler R. Endothelial progenitor cells: Exploring the pleiotropic effects of statins. World J Cardiol 2017; 9:1-13. [PMID: 28163831 PMCID: PMC5253189 DOI: 10.4330/wjc.v9.i1.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/29/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023] Open
Abstract
Statins have become a cornerstone of risk modification for ischaemic heart disease patients. A number of studies have shown that they are effective and safe. However studies have observed an early benefit in terms of a reduction in recurrent infarct and or death after a myocardial infarction, prior to any significant change in lipid profile. Therefore, pleiotropic mechanisms, other than lowering lipid profile alone, must account for this effect. One such proposed pleiotropic mechanism is the ability of statins to augment both number and function of endothelial progenitor cells. The ability to augment repair and maintenance of a functioning endothelium may have profound beneficial effect on vascular repair and potentially a positive impact on clinical outcomes in patients with cardiovascular disease. The following literature review will discuss issues surrounding endothelial progenitor cell (EPC) identification, role in vascular repair, factors affecting EPC numbers, the role of statins in current medical practice and their effects on EPC number.
Collapse
|
27
|
Metabolic hijacking: A survival strategy cancer cells exploit? Crit Rev Oncol Hematol 2017; 109:1-8. [DOI: 10.1016/j.critrevonc.2016.11.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022] Open
|
28
|
Özcan B, Leenen PJM, Delhanty PJD, Baldéon-Rojas LY, Neggers SJ, van der Lely AJ. Unacylated ghrelin modulates circulating angiogenic cell number in insulin-resistant states. Diabetol Metab Syndr 2017; 9:43. [PMID: 28572856 PMCID: PMC5452348 DOI: 10.1186/s13098-017-0239-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 05/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is associated with reduced numbers and impaired function of circulating angiogenic cells (CAC) which contributes to the progression of atherosclerosis and microvascular disease. Previous studies suggest that short-term infusion of unacylated ghrelin (UAG) normalizes CAC number in patients with T2D. To determine dose-dependent effects of short-term infusion of UAG in T2D patients using a cross-over model, and of long-term infusion of UAG in obese mice, on differentiation of monocyte progenitors into CAC. METHODS Eight overweight T2D patients were infused overnight with 3 and 10 µg/kg/h of UAG in a double-blind, placebo-controlled cross-over study. To assess the effects of long-term UAG treatment, obese mice were infused with UAG for 4 weeks. Monocyte progenitors were assessed for their ability to differentiate into CAC in vitro. RESULTS In T2D patients, UAG treatment caused a reduction in differentiation of CAC, dependent on UAG dose and differentiation method. However, mice treated with UAG showed a significant increase in differentiation of bone marrow progenitors into CAC. CONCLUSION UAG causes a minor suppressive effect on CAC development after short-term treatment in humans, but experiments in mice suggest that long-term treatment has beneficial effects on CAC formation. The Netherlands Trial Register: TC=2487.
Collapse
Affiliation(s)
- Behiye Özcan
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
29
|
Atorvastatin enhances endothelial cell function in posttransplant poor graft function. Blood 2016; 128:2988-2999. [PMID: 27769957 DOI: 10.1182/blood-2016-03-702803] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
Key Points
Dysfunctional BM EPCs were found in subjects with PGF postallotransplant. BM EPCs from subjects with PGF were enhanced by atorvastatin through downregulation of the p38 MAPK pathway.
Collapse
|
30
|
Kumar S, Batra A, Kanthaje S, Ghosh S, Chakraborti A. Crosstalk between microRNA-122 and FOX family genes in HepG2 cells. Exp Biol Med (Maywood) 2016; 242:436-440. [PMID: 27895094 DOI: 10.1177/1535370216681548] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
MicroRNA-122 (miR-122) is liver specific and plays an important role in physiology as well as diseases including hepatocellular carcinoma (HCC). Downregulation of miR-122 in HCC modulates apoptosis. Similarly, the putative targets of miR-122, the forkhead box (FOX) family genes also play an important role in the regulation of apoptosis. Hence, an interplay between miR-122 and FOX family genes has been explored in this study. Initially, an augmentation of apoptosis was noticed in HepG2 cells after transfection with miR-122. Further, the predicted miR-122 targets, the FOX family genes ( FOXM1b, FOXP1, and FOXO4) were selected via in silico analysis based on their role in apoptosis. We checked the expression of all these genes at transcript level after the transfection of miR-122 and found that the relative expression of FOXP1 and FOXM1b was significantly downregulated (p < 0.005) and that of FOXO4 was upregulated (p < 0.005). Thus, the finding indicates deregulation of these FOX genes as a result of miR-122 augmentation might be involved in the modulation of apoptosis.
Collapse
Affiliation(s)
- Subodh Kumar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ankita Batra
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Shruthi Kanthaje
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Sujata Ghosh
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Anuradha Chakraborti
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
31
|
Xu Y, Zhu J, Hu X, Wang C, Lu D, Gong C, Yang J, Zong L. CLIC1 Inhibition Attenuates Vascular Inflammation, Oxidative Stress, and Endothelial Injury. PLoS One 2016; 11:e0166790. [PMID: 27861612 PMCID: PMC5115793 DOI: 10.1371/journal.pone.0166790] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 11/03/2016] [Indexed: 01/26/2023] Open
Abstract
Endothelial dysfunction, which includes endothelial oxidative damage and vascular inflammation, is a key initiating step in the pathogenesis of atherosclerosis (AS) and an independent risk factor for this disorder. Intracellular chloride channel 1 (CLIC1), a novel metamorphic protein, acts as a sensor of cell oxidation and is involved in inflammation. In this study, we hypothesize that CLIC1 plays an important role in AS. Apolipoprotein E-deficient mice were supplied with a normal diet or a high-fat and high-cholesterol diet for 8 weeks. Overexpressed CLIC1 was associated with the accelerated atherosclerotic plaque development, amplified oxidative stress, and in vivo release of inflammatory cytokines. We subsequently examined the underlying molecular mechanisms through in vitro experiments. Treatment of cultured human umbilical vein endothelial cells (HUVECs) with H2O2 induced endothelial oxidative damage and enhanced CLIC1 expression. Suppressing CLIC1 expression through gene knocked-out (CLIC1-/-) or using the specific inhibitor indanyloxyacetic acid-94 (IAA94) reduced ROS production, increased SOD enzyme activity, and significantly decreased MDA level. CLIC1-/- HUVECs exhibited significantly reduced expression of TNF-α and IL-1β as well as ICAM-1 and VCAM-1 at the protein levels. In addition, H2O2 promoted CLIC1 translocation to the cell membrane and insertion into lipid membranes, whereas IAA94 inhibited CLIC1 membrane translocation induced by H2O2. By contrast, the majority of CLIC1 did not aggregate on the cell membrane in normal HUVECs, and this finding is consistent with the changes in cytoplasmic chloride ion concentration. This study demonstrates for the first time that CLIC1 is overexpressed during AS development both in vitro and in vivo and can regulate the accumulation of inflammatory cytokines and production of oxidative stress. Our results also highlight that deregulation of endothelial functions may be associated with the membrane translocation of CLIC1 and active chloride-selective ion channels in endothelial cells.
Collapse
Affiliation(s)
- Yingling Xu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ji Zhu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao Hu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cui Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dezhao Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
- * E-mail:
| | - Chenxue Gong
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinhuan Yang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei Zong
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Yan YH, Li SH, Li HY, Lin Y, Yang JX. Osthole Protects Bone Marrow-Derived Neural Stem Cells from Oxidative Damage through PI3K/Akt-1 Pathway. Neurochem Res 2016; 42:398-405. [DOI: 10.1007/s11064-016-2082-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/30/2016] [Accepted: 10/06/2016] [Indexed: 12/13/2022]
|
33
|
Wu K, Ma J, Bai W, Cui X, Han T, Wang S, Xie Y, Xie Y. Short-term intratracheal use of PEG-modified IL-2 and glucocorticoid persistently alleviates asthma in a mouse model. Sci Rep 2016; 6:31562. [PMID: 27527926 PMCID: PMC4985708 DOI: 10.1038/srep31562] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/22/2016] [Indexed: 01/12/2023] Open
Abstract
Regulatory T (Treg) cells play an important role in allergic airway diseases, and upregulation of Treg cells is a potential therapeutic strategy for asthma. In this study, we show that short-term intratracheal use of IL-2 combined with glucocorticoid alleviates antigen-induced airway inflammation and reduces airway hyperresponsiveness by expanding antigen-nonspecific Treg cells, with a decrease in T helper 2 (Th2) cells and Th2-associated cytokines. We also designed a long-acting polyethylene glycol (PEG)-modified IL-2 and demonstrated that the optimal dosage form is IL-2(PEG) plus budesonide, which can upregulate Treg cells and ameliorate asthma at a lower dose. The therapeutic effect was faster than treatment with dexamethasone and was effective at a low dose suitable for humans that could last for at least 6 weeks. This study unveils a new therapeutic regimen and suggests that such endogenous Treg therapy could be a useful tool to persistently alleviate asthma.
Collapse
Affiliation(s)
- Kefei Wu
- Department of Hematology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, People's Republic of China
| | - Jiexian Ma
- Department of Hematology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, People's Republic of China
| | - Weiya Bai
- Key laboratory of medical molecular virology, Institutes of biomedical sciences and institute of medical microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoxian Cui
- Key laboratory of medical molecular virology, Institutes of biomedical sciences and institute of medical microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Tao Han
- Department of Hematology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, People's Republic of China
| | - Shiyuan Wang
- Xiamen Amoytop Biotech Co., Ltd, Xiamen 360000, People's Republic of China
| | - Youhua Xie
- Key laboratory of medical molecular virology, Institutes of biomedical sciences and institute of medical microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yanhui Xie
- Department of Hematology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, People's Republic of China
| |
Collapse
|
34
|
Jantzen K, Møller P, Karottki DG, Olsen Y, Bekö G, Clausen G, Hersoug LG, Loft S. Exposure to ultrafine particles, intracellular production of reactive oxygen species in leukocytes and altered levels of endothelial progenitor cells. Toxicology 2016; 359-360:11-8. [PMID: 27311922 DOI: 10.1016/j.tox.2016.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022]
Abstract
Exposure to particles in the fine and ultrafine size range has been linked to induction of low-grade systemic inflammation, oxidative stress and development of cardiovascular diseases. Declining levels of endothelial progenitor cells within systemic circulation have likewise been linked to progression of cardiovascular diseases. The objective was to determine if exposure to fine and ultrafine particles from indoor and outdoor sources, assessed by personal and residential indoor monitoring, is associated with altered levels of endothelial progenitor cells, and whether such effects are related to leukocyte-mediated oxidative stress. The study utilized a cross sectional design performed in 58 study participants from a larger cohort. Levels of circulating endothelial progenitor cells, defined as either late (CD34(+)KDR(+) cells) or early (CD34(+)CD133(+)KDR(+) cells) subsets were measured using polychromatic flow cytometry. We additionally measured production of reactive oxygen species in leukocyte subsets (lymphocytes, monocytes and granulocytes) by flow cytometry using intracellular 2',7'-dichlorofluoroscein. The measurements encompassed both basal levels of reactive oxygen species production and capacity for reactive oxygen species production for each leukocyte subset. We found that the late endothelial progenitor subset was negatively associated with levels of ultrafine particles measured within the participant residences and with reactive oxygen species production capacity in lymphocytes. Additionally, the early endothelial progenitor cell levels were positively associated with a personalised measure of ultrafine particle exposure and negatively associated with both basal and capacity for reactive oxygen species production in lymphocytes and granulocytes, respectively. Our results indicate that exposure to fine and ultrafine particles derived from indoor sources may have adverse effects on human vascular health.
Collapse
Affiliation(s)
- Kim Jantzen
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Peter Møller
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorina Gabriela Karottki
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yulia Olsen
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gabriel Bekö
- International Centre for Indoor Environment and Energy, Department of Civil Engineering, Technical University of Denmark, Lyngby, Denmark
| | - Geo Clausen
- International Centre for Indoor Environment and Energy, Department of Civil Engineering, Technical University of Denmark, Lyngby, Denmark
| | - Lars-Georg Hersoug
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Loft
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Ning Y, Li Z, Qiu Z. FOXO1 silence aggravates oxidative stress-promoted apoptosis in cardiomyocytes by reducing autophagy. J Toxicol Sci 2016; 40:637-45. [PMID: 26354380 DOI: 10.2131/jts.40.637] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Mechanisms underlining oxidative stress-induced injury to cardiomyocytes during myocardial infarction (MI) or acute ischemia/reperfusion (I/R) are not well recognized. Forkhead box O (FOXO) transcription factors have been defined as critical mediators of oxidative stress resistance in multiple cell types, but their cardioprotective functions have not been reported previously. In the present study, we investigated the promotion to FOXO1 by the treatment with hydrogen peroxide (H2O2) during the H2O2-induced apoptosis in cardiomyocyte H9c2 cells. We then silenced FOXO1 with FOXO1-specific siRNA, and re-evaluated the H2O2-induced apoptosis. In addition, we also examined the H2O2-induced autophagy and the autophagy induction post FOXO1 silence. Results demonstrated that H2O2 induced a significantly high level of apoptosis in H9c2 cells. Interestingly, the FOXO1 in both mRNA and protein levels were not significantly regulated, however, the phosphorylated form of FOXO1 was significantly promoted in the H2O2-treated H9c2 cells. On the other hand, post the significant knockout of FOXO1 with the transfection with FOXO1-specific siRNA, the apoptosis induction was more significant in H9c2 cells subjected to H2O2. In addition, we found a significantly higher level of autophagy induction in the H2O2-treated H9c2 cells. However, the autophagy was markedly reduced by the knockout of FOXO1. In summary, these data support the critical role for FOXO1 in promoting cardiomyocytes against oxidative stress probably through inducing autophagy.
Collapse
Affiliation(s)
- Yuzhen Ning
- Vasculocardiology Deparment, Zhujiang Hospital, Southern Medical University, China
| | | | | |
Collapse
|
36
|
Wang YW, Zhang JH, Yu Y, Yu J, Huang L. Inhibition of Store-Operated Calcium Entry Protects Endothelial Progenitor Cells from H2O2-Induced Apoptosis. Biomol Ther (Seoul) 2016; 24:371-9. [PMID: 27169819 PMCID: PMC4930280 DOI: 10.4062/biomolther.2015.130] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/09/2015] [Accepted: 11/17/2015] [Indexed: 01/09/2023] Open
Abstract
Store-operated calcium entry (SOCE), a major mode of extracellular calcium entry, plays roles in a variety of cell activities. Accumulating evidence indicates that the intracellular calcium ion concentration and calcium signaling are critical for the responses induced by oxidative stress. The present study was designed to investigate the potential effect of SOCE inhibition on H2O2-induced apoptosis in endothelial progenitor cells (EPCs), which are the predominant cells involved in endothelial repair. The results showed that H2O2-induced EPC apoptosis was reversed by SOCE inhibition induced either using the SOCE antagonist ML-9 or via silencing of stromal interaction molecule 1 (STIM1), a component of SOCE. Furthermore, SOCE inhibition repressed the increases in intracellular reactive oxygen species (ROS) levels and endoplasmic reticulum (ER) stress and ameliorated the mitochondrial dysfunction caused by H2O2. Our findings provide evidence that SOCE inhibition exerts a protective effect on EPCs in response to oxidative stress induced by H2O2 and may serve as a potential therapeutic strategy against vascular endothelial injury.
Collapse
Affiliation(s)
- Yan-Wei Wang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Ji-Hang Zhang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Yang Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Jie Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| | - Lan Huang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, People's Republic of China
| |
Collapse
|
37
|
Wu JR, Hsu JH, Dai ZK, Wu BN, Chen IJ, Liou SF, Yeh JL. Activation of endothelial NO synthase by a xanthine derivative ameliorates hypoxia-induced apoptosis in endothelial progenitor cells. ACTA ACUST UNITED AC 2016; 68:810-8. [PMID: 27109251 DOI: 10.1111/jphp.12555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 03/13/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Endothelial damage is strongly associated with cardiovascular diseases such as atherosclerosis, thrombosis and hypertension. Endothelial progenitor cells (EPCs) are primitive bone marrow (BM) cells that possess the capacity to mature into endothelial cells and play a role in neovascularization and vascular remodelling. This study aimed to investigate whether KMUP-1, a synthetic xanthine-based derivative, atorvastatin and simvastatin, can prevent endothelial dysfunction and apoptosis induced by hypoxia and to elucidate the underlying mechanisms. METHODS Mononuclear cells were separated and were induced to differentiate into EPCs. KMUP-1, atorvastatin or simvastatin were administered prior to hypoxia. KEY FINDINGS We found that EPCs exposed to hypoxia increased apoptosis as well as diminished proliferation. Pretreatment with KMUP-1, atorvastatin and simvastatin significantly prevented hypoxia-induced EPCs death and apoptosis, with associated increased of the Bcl-2/Bax ratio, and reduced caspase-3 and caspase-9 expression. We also assessed the nitrite production and Ser(1177)-phospho-eNOS expression and found that KMUP-1, atorvastatin and simvastatin not only increased the secretion of NO compared with the hypoxia group but also upregulated the eNOS activation. CONCLUSIONS KMUP-1 inhibited hypoxia-induced dysfunction and apoptosis in EPCs, which may be mediated through suppressing oxidative stress, upregulating eNOS and downregulating the caspase-3 signalling pathway.
Collapse
Affiliation(s)
- Jiunn-Ren Wu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zen-Kong Dai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Paediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ing-Jun Chen
- Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Fen Liou
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Jwu-Lai Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department and Graduate Institute of Pharmacology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
38
|
4EBP1/c-MYC/PUMA and NF-κB/EGR1/BIM pathways underlie cytotoxicity of mTOR dual inhibitors in malignant lymphoid cells. Blood 2016; 127:2711-22. [PMID: 26917778 DOI: 10.1182/blood-2015-02-629485] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 02/13/2016] [Indexed: 12/14/2022] Open
Abstract
The mammalian target of rapamycin (mTOR), a kinase that regulates proliferation and apoptosis, has been extensively evaluated as a therapeutic target in multiple malignancies. Rapamycin analogs, which partially inhibit mTOR complex 1 (mTORC1), exhibit immunosuppressive and limited antitumor activity, but sometimes activate survival pathways through feedback mechanisms involving mTORC2. Thus, attention has turned to agents targeting both mTOR complexes by binding the mTOR active site. Here we show that disruption of either mTOR-containing complex is toxic to acute lymphocytic leukemia (ALL) cells and identify 2 previously unrecognized pathways leading to this cell death. Inhibition of mTORC1-mediated 4EBP1 phosphorylation leads to decreased expression of c-MYC and subsequent upregulation of the proapoptotic BCL2 family member PUMA, whereas inhibition of mTORC2 results in nuclear factor-κB-mediated expression of the Early Growth Response 1 (EGR1) gene, which encodes a transcription factor that binds and transactivates the proapoptotic BCL2L11 locus encoding BIM. Importantly, 1 or both pathways contribute to death of malignant lymphoid cells after treatment with dual mTORC1/mTORC2 inhibitors. Collectively, these observations not only provide new insight into the survival roles of mTOR in lymphoid malignancies, but also identify alterations that potentially modulate the action of mTOR dual inhibitors in ALL.
Collapse
|
39
|
WANG WEI, ZHOU PANGHU, HU WEI. Overexpression of FOXO4 induces apoptosis of clear-cell renal carcinoma cells through downregulation of Bim. Mol Med Rep 2016; 13:2229-34. [DOI: 10.3892/mmr.2016.4789] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 10/22/2015] [Indexed: 11/06/2022] Open
|
40
|
Kanzaki H, Shinohara F, Kanako I, Yamaguchi Y, Fukaya S, Miyamoto Y, Wada S, Nakamura Y. Molecular regulatory mechanisms of osteoclastogenesis through cytoprotective enzymes. Redox Biol 2016; 8:186-91. [PMID: 26795736 PMCID: PMC4732015 DOI: 10.1016/j.redox.2016.01.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 12/30/2022] Open
Abstract
It has been reported that reactive oxygen species (ROS), such as hydrogen peroxide and superoxide, take part in osteoclast differentiation as intra-cellular signaling molecules. The current assumed signaling cascade from RANK to ROS production is RANK, TRAF6, Rac1, and then Nox. The target molecules of ROS in RANKL signaling remain unclear; however, several reports support the theory that NF-κB signaling could be the crucial downstream signaling molecule of RANKL-mediated ROS signaling. Furthermore, ROS exert cytotoxic effects such as peroxidation of lipids and phospholipids and oxidative damage to proteins and DNA. Therefore, cells have several protective mechanisms against oxidative stressors that mainly induce cytoprotective enzymes and ROS scavenging. Three well-known mechanisms regulate cytoprotective enzymes including Nrf2-, FOXO-, and sirtuin-dependent mechanisms. Several reports have indicated a crosslink between FOXO- and sirtuin-dependent regulatory mechanisms. The agonists against the regulatory mechanisms are reported to induce these cytoprotective enzymes successfully. Some of them inhibit osteoclast differentiation and bone destruction via attenuation of intracellular ROS signaling. In this review article, we discuss the above topics and summarize the current information available on the relationship between cytoprotective enzymes and osteoclastogenesis.
Collapse
Affiliation(s)
- Hiroyuki Kanzaki
- Tohoku University Hospital, Maxillo-Oral Disorders, Japan; Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan.
| | - Fumiaki Shinohara
- Tohoku University Graduate School of Dentistry, Oral Microbiology, Japan
| | - Itohiya Kanako
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| | - Yuuki Yamaguchi
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| | - Sari Fukaya
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| | - Yutaka Miyamoto
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| | - Satoshi Wada
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| | - Yoshiki Nakamura
- Department of orthodontics, School of Dental Medicine, Tsurumi University, Japan
| |
Collapse
|
41
|
Abstract
In multicellular organisms, cell death is a critical and active process that maintains tissue homeostasis and eliminates potentially harmful cells. There are three major types of morphologically distinct cell death: apoptosis (type I cell death), autophagic cell death (type II), and necrosis (type III). All three can be executed through distinct, and sometimes overlapping, signaling pathways that are engaged in response to specific stimuli. Apoptosis is triggered when cell-surface death receptors such as Fas are bound by their ligands (the extrinsic pathway) or when Bcl2-family proapoptotic proteins cause the permeabilization of the mitochondrial outer membrane (the intrinsic pathway). Both pathways converge on the activation of the caspase protease family, which is ultimately responsible for the dismantling of the cell. Autophagy defines a catabolic process in which parts of the cytosol and specific organelles are engulfed by a double-membrane structure, known as the autophagosome, and eventually degraded. Autophagy is mostly a survival mechanism; nevertheless, there are a few examples of autophagic cell death in which components of the autophagic signaling pathway actively promote cell death. Necrotic cell death is characterized by the rapid loss of plasma membrane integrity. This form of cell death can result from active signaling pathways, the best characterized of which is dependent on the activity of the protein kinase RIP3.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Fabien Llambi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
42
|
Demolli S, Doebele C, Doddaballapur A, Lang V, Fisslthaler B, Chavakis E, Vinciguerra M, Sciacca S, Henschler R, Hecker M, Savant S, Augustin HG, Kaluza D, Dimmeler S, Boon RA. MicroRNA-30 mediates anti-inflammatory effects of shear stress and KLF2 via repression of angiopoietin 2. J Mol Cell Cardiol 2015; 88:111-9. [PMID: 26456066 DOI: 10.1016/j.yjmcc.2015.10.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 01/13/2023]
Abstract
MicroRNAs are endogenously expressed small noncoding RNAs that regulate gene expression. Laminar blood flow induces atheroprotective gene expression in endothelial cells (ECs) in part by upregulating the transcription factor KLF2. Here, we identified KLF2- and flow-responsive miRs that affect gene expression in ECs. Bioinformatic assessment of mRNA expression patterns identified the miR-30-5p seed sequence to be highly enriched in mRNAs that are downregulated by KLF2. Indeed, KLF2 overexpression and shear stress stimulation in vitro and in vivo increased the expression of miR-30-5p family members. Furthermore, we identified angiopoietin 2 (Ang2) as a target of miR-30. MiR-30 overexpression reduces Ang2 levels, whereas miR-30 inhibition by LNA-antimiRs induces Ang2 expression. Consistently, miR-30 reduced basal and TNF-α-induced expression of the inflammatory cell–cell adhesion molecules E-selectin, ICAM1 and VCAM1, which was rescued by stimulation with exogenous Ang2. In summary, KLF2 and shear stress increase the expression of the miR-30-5p family which acts in an anti-inflammatory manner in ECs by impairing the expression of Ang2 and inflammatory cell–cell adhesion molecules. The upregulation of miR-30-5p family members may contribute to the atheroprotective effects of shear stress.
Collapse
Affiliation(s)
- Shemsi Demolli
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Carmen Doebele
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Anuradha Doddaballapur
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Victoria Lang
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen GmbH, Frankfurt, Germany
| | - Beate Fisslthaler
- Institute for Vascular Signaling, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Emmanouil Chavakis
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; Division of Internal Medicine III, Cardiology, Hospital of the Goethe University, Frankfurt, Germany
| | | | - Sergio Sciacca
- Cardiac Surgery and Heart Transplantation Unit, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Reinhard Henschler
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen GmbH, Frankfurt, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Soniya Savant
- Medical Faculty Mannheim (CBTM), Heidelberg University, Germany
| | - Hellmut G Augustin
- Medical Faculty Mannheim (CBTM), Heidelberg University, Germany; German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - David Kaluza
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany..
| |
Collapse
|
43
|
Wang WR, Liu EQ, Zhang JY, Li YX, Yang XF, He YH, Zhang W, Jing T, Lin R. Activation of PPAR alpha by fenofibrate inhibits apoptosis in vascular adventitial fibroblasts partly through SIRT1-mediated deacetylation of FoxO1. Exp Cell Res 2015; 338:54-63. [DOI: 10.1016/j.yexcr.2015.07.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/09/2015] [Accepted: 07/25/2015] [Indexed: 11/30/2022]
|
44
|
Sionov RV, Vlahopoulos SA, Granot Z. Regulation of Bim in Health and Disease. Oncotarget 2015; 6:23058-134. [PMID: 26405162 PMCID: PMC4695108 DOI: 10.18632/oncotarget.5492] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/08/2015] [Indexed: 11/25/2022] Open
Abstract
The BH3-only Bim protein is a major determinant for initiating the intrinsic apoptotic pathway under both physiological and pathophysiological conditions. Tight regulation of its expression and activity at the transcriptional, translational and post-translational levels together with the induction of alternatively spliced isoforms with different pro-apoptotic potential, ensure timely activation of Bim. Under physiological conditions, Bim is essential for shaping immune responses where its absence promotes autoimmunity, while too early Bim induction eliminates cytotoxic T cells prematurely, resulting in chronic inflammation and tumor progression. Enhanced Bim induction in neurons causes neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Moreover, type I diabetes is promoted by genetically predisposed elevation of Bim in β-cells. On the contrary, cancer cells have developed mechanisms that suppress Bim expression necessary for tumor progression and metastasis. This review focuses on the intricate network regulating Bim activity and its involvement in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| | - Spiros A. Vlahopoulos
- First Department of Pediatrics, University of Athens, Horemeio Research Laboratory, Thivon and Levadias, Goudi, Athens, Greece
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel Canada, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
45
|
Lupieri A, Smirnova N, Malet N, Gayral S, Laffargue M. PI3K signaling in arterial diseases: Non redundant functions of the PI3K isoforms. Adv Biol Regul 2015; 59:4-18. [PMID: 26238239 DOI: 10.1016/j.jbior.2015.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 06/04/2023]
Abstract
Cardiovascular diseases are the most common cause of death around the world. This includes atherosclerosis and the adverse effects of its treatment, such as restenosis and thrombotic complications. The development of these arterial pathologies requires a series of highly-intertwined interactions between immune and arterial cells, leading to specific inflammatory and fibroproliferative cellular responses. In the last few years, the study of phosphoinositide 3-kinase (PI3K) functions has become an attractive area of investigation in the field of arterial diseases, especially since inhibitors of specific PI3K isoforms have been developed. The PI3K family includes 8 members divided into classes I, II or III depending on their substrate specificity. Although some of the different isoforms are responsible for the production of the same 3-phosphoinositides, they each have specific, non-redundant functions as a result of differences in expression levels in different cell types, activation mechanisms and specific subcellular locations. This review will focus on the functions of the different PI3K isoforms that are suspected as having protective or deleterious effects in both the various immune cells and types of cell found in the arterial wall. It will also discuss our current understanding in the context of which PI3K isoform(s) should be targeted for future therapeutic interventions to prevent or treat arterial diseases.
Collapse
Affiliation(s)
- Adrien Lupieri
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Natalia Smirnova
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Nicole Malet
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France.
| |
Collapse
|
46
|
Xu ZH, Shun WW, Hang JB, Gao BL, Hu JA. Posttranslational modifications of FOXO1 regulate epidermal growth factor receptor tyrosine kinase inhibitor resistance for non-small cell lung cancer cells. Tumour Biol 2015; 36:5485-95. [PMID: 26036758 DOI: 10.1007/s13277-015-3215-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 02/03/2015] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (TKIs) are effective clinical therapies for advanced non-small cell lung cancer (NSCLC) patients, while resistance to TKIs remains a serious problem in clinical practice. Recently, it has been proposed that targeting mTOR could overcome TKI resistance in NSCLC cells. Forkhead box class O1 (FOXO1) has emerged as an important rheostat that modulates the activity of Akt and mTOR signaling pathway. However, the role of FOXO1 and related regulatory mechanism in TKI resistance in NSCLC remain largely unknown. Here, we find that mTOR-AKT-FOXO1 signaling cascade is deregulated in TKI-resistant NSCLC cells and that FOXO1 was highly phosphorylated and lowly acetylated upon erlotinib treatment. Combination of mTOR or PI3K inhibitor and erlotinib overcomes TKI resistance to inhibit cell growth and induce apoptosis in TKI-resistant NSCLC cells. Furthermore, the phosphorylation and acetylation of FOXO1 are reversely modulated by mTORC2-AKT signaling pathway. FOXO1 mutation analyses reveal that FOXO1 acetylation inhibits cell proliferation and promotes NSCLC cell apoptosis, while the phosphorylation of FOXO1 plays opposite roles in NSCLC cells. Importantly, increasing FOXO1 acetylation by a HDAC inhibitor, depsipeptide, overcomes TKI resistance to effectively induce TKI-resistant NSCLC cell apoptosis. Together, FOXO1 plays dual roles in TKI resistance through posttranslational modifications in NSCLC and this study provides a possible strategy for treatment of TKI-resistant NSCLC patients.
Collapse
Affiliation(s)
- Zhi-hong Xu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | | | | | | | | |
Collapse
|
47
|
Ono YJ, Tanabe A, Nakamura Y, Yamamoto H, Hayashi A, Tanaka T, Sasaki H, Hayashi M, Terai Y, Ohmichi M. A low-testosterone state associated with endometrioma leads to the apoptosis of granulosa cells. PLoS One 2014; 9:e115618. [PMID: 25536335 PMCID: PMC4275210 DOI: 10.1371/journal.pone.0115618] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/27/2014] [Indexed: 01/15/2023] Open
Abstract
Although endometriosis is suspected to be a cause of premature ovarian insufficiency (POI), the mechanism(s) underlying this process have not been elucidated. Recently, androgens were shown to promote oocyte maturation and to play a role in folliculogenesis. In addition, several reports have documented low testosterone levels in the follicular fluid obtained from endometriosis patients. We therefore examined whether the low levels of serum testosterone are associated with the apoptosis of granulosa cells in follicles obtained from endometriosis patients. Serum samples were collected from 46 patients with endometriosis and from 62 patients without endometriosis who received assisted reproductive therapy. Specimens of the ovaries obtained from 10 patients with endometrioma were collected using laparoscopy. The mean serum testosterone concentration in the patients with endometriosis was significantly lower than that observed in the patients without endometriosis. Furthermore, high expression of a pro-apoptotic Bcl-2 member, BimEL, in the follicles was found to be associated with a low serum testosterone level. We clarified the underlying mechanisms using a basic approach employing human immortalized granulosa cells derived from a primary human granulosa cell tumor, the COV434 cell line. The in vitro examination demonstrated that testosterone inhibited apoptosis induced by sex steroids depletion via the PI3K/Akt-FoxO3a pathway in the COV434 cells. In conclusion, we elucidated the mechanism underlying the anti-apoptotic effects of testosterone on granulosa cells, and found that a low-testosterone status is a potentially important step in the development of premature ovarian insufficiency in patients with endometriosis.
Collapse
Affiliation(s)
- Yoshihiro J. Ono
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Akiko Tanabe
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
- * E-mail:
| | - Yoko Nakamura
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Hikaru Yamamoto
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Atsushi Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Hiroshi Sasaki
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Masami Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| |
Collapse
|
48
|
Huang C, Li J, Hong K, Xia Z, Xu Y, Cheng X. BH3-only protein Bim is upregulated and mediates the apoptosis of cardiomyocytes under glucose and oxygen-deprivation conditions. Cell Biol Int 2014; 39:318-25. [PMID: 25319047 DOI: 10.1002/cbin.10392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 09/12/2014] [Indexed: 12/31/2022]
Abstract
Bim is a potent pro-apoptotic BH3-only Bcl-2 member. However, the expression of Bim and its role in cardiac injury induced by ischemia remain unclear. H9c2 cells were subjected to a glucose and oxygen-deprived (GOD) condition in vitro, mimicking ischemia environment in vivo. GOD treatment augmented the expression of Bim and induced the apoptosis of H9c2 cells. Silencing of Bim by RNAi significantly attenuated GOD-induced cytotoxicity, suppressed mitochondrial membrane potential △Ψm loss, inhibited caspase 3 activation and reduced apoptosis. The data demonstrate that Bim is upregulated by GOD in a time-dependent manner in H9c2 cells, and enhances mitochondrial apoptosis dependent on the activation of caspase 3. Silencing of Bim may be a promising therapeutic strategy in ischemia related heart diseases.
Collapse
Affiliation(s)
- Chahua Huang
- Department of Cardiology, Second affiliated hospital, Nanchang University, Nanchang, 330006, China
| | | | | | | | | | | |
Collapse
|
49
|
Involvement of FoxO1 in the effects of follicle-stimulating hormone on inhibition of apoptosis in mouse granulosa cells. Cell Death Dis 2014; 5:e1475. [PMID: 25321482 PMCID: PMC4237239 DOI: 10.1038/cddis.2014.400] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/23/2022]
Abstract
In mammalian ovaries, follicular atresia occurs periodically and destroys almost all the follicles in the ovary. Follicle-stimulating hormone (FSH) acts as the primary survival factor during follicular atresia by preventing apoptosis in granulosa cells. FoxO1 is a critical factor in promoting follicular atresia and granulosa cell apoptosis. FSH inhibits the induction of FoxO1. In this report, we investigated the role of FSH-FoxO1 pathway in mouse follicular atresia. FSH dampened stress-induced apoptosis and the expression of FoxO1 and pro-apoptosis genes in mouse granulosa cells (MGCs). In contrast, overexpression of FoxO1 inhibited the viability of MGCs and induced the expression of endogenous FoxO1. The signaling cascades involved in regulating FoxO1 activity upon FSH treatment were identified using FSH signaling antagonists. Blocking protein kinase A (PKA), phosphatidylinositol-3 kinase (PI3K) or protein kinase B (AKT) restored the upregulation of FoxO1 and apoptotic signals, which was suppressed by FSH. Moreover, inhibition of PKA or PI3K impaired FSH-induced AKT activity, but inactivation of PI3K or AKT had little effect on PKA activity in the presence of FSH. Correspondingly, constitutive activation of FoxO1 (all three AKT sites were replaced by alanines) also promoted MGC apoptosis despite FSH administration. Furthermore, both luciferase reporter assays and chromatin immunoprecipitation assays showed that FoxO1 directly bound to a FoxO-recognized element site within the FoxO1 promoter and contributed to the regulation of FoxO1 expression in response to FSH. Taken together, we propose a novel model in which FSH downregulates FoxO1-dependent apoptosis in MGCs by coordinating the PKA-PI3K-AKT-FoxO1 axis and FoxO1-FoxO1 positive feedback.
Collapse
|
50
|
Abstract
Endothelial progenitor cells (EPCs) are primitive endothelial precursors which are known to functionally contribute to the pathogenesis of disease. To date a number of distinct subtypes of these cells have been described, with differing maturation status, cellular phenotype, and function. Although there is much debate on which subtype constitutes the true EPC population, all subtypes have endothelial characteristics and contribute to neovascularisation. Vasculogenesis, the process by which EPCs contribute to blood vessel formation, can be dysregulated in disease with overabundant vasculogenesis in the context of solid tumours, leading to tumour growth and metastasis, and conversely insufficient vasculogenesis can be present in an ischemic environment. Importantly, it is widely known that transcription factors tightly regulate cellular phenotype and function by controlling the expression of particular target genes and in turn regulating specific signalling pathways. This suggests that transcriptional regulators may be potential therapeutic targets to control EPC function. Herein, we discuss the observed EPC subtypes described in the literature and review recent studies describing the role of a number of transcriptional families in the regulation of EPC phenotype and function in normal and pathological conditions.
Collapse
|