101
|
Lu X, Wang Y, Zhang J, Pu D, Hu N, Luo J, An Q, He L. Patients with systemic lupus erythematosus face a high risk of cardiovascular disease: A systematic review and Meta-analysis. Int Immunopharmacol 2021; 94:107466. [PMID: 33636561 DOI: 10.1016/j.intimp.2021.107466] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/24/2021] [Accepted: 02/01/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) have increased mortality related to cardiovascular disease (CVD). This systematic review and meta-analysis identified the risk of CVD in SLE patients, CVD risk factors in SLE patients, and the risk of CVD in lupus nephritis (LN) patients. METHODS On-line databases were used to search the eligible studies from January 2013 to August 2020. The relevant characteristics and the data of disease extracted from included publications. RESULTS A total of 20 studies were included in this meta-analysis. Compared with the general or healthy population, the risk of CVD in SLE patients increased by 2 times (RR = 2.35, 95% CI: 1.95-2.84, P < 0.05). SLE patients had a significantly increased risk of atherosclerosis (RR = 2.31, 95% CI: 1.16-4.60), stroke (RR = 2.30, 95% CI: 1.52-3.50), myocardial infarction (RR = 2.66, 95% CI: 1.97-3.59), peripheral vascular disease (RR = 2.56, 95% CI: 1.07-6.09) and heart failure (RR = 2.89, 95% CI: 1.63-5.13), but no significant increased risk of coronary artery disease (RR = 1.93, 95% CI: 0.67-5.59). SLE patients were more susceptible to lead hypertension than general or healthy population (RR = 2.31, 95% CI: 1.62-3.29). Compared with the SLE patients, the risk of CVD in LN patients was increased by 2 times (RR = 1.75, 95% CI: 1.13-2.70). CONCLUSION The results of this meta-analysis suggest that SLE patients have a higher risk of developing CVD compared with the general or healthy population, and the risk of CVD in LN patients is significantly higher than that in SLE patients.
Collapse
Affiliation(s)
- Xiaohong Lu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - YanHua Wang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Zhang
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dan Pu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Nan Hu
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Luo
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi An
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lan He
- Department of Rheumatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
102
|
Sillen M, Miyata T, Vaughan DE, Strelkov SV, Declerck PJ. Structural Insight into the Two-Step Mechanism of PAI-1 Inhibition by Small Molecule TM5484. Int J Mol Sci 2021; 22:ijms22031482. [PMID: 33540702 PMCID: PMC7867230 DOI: 10.3390/ijms22031482] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 01/19/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1), a key regulator of the fibrinolytic system, is the main physiological inhibitor of plasminogen activators. By interacting with matrix components, including vitronectin (Vn), PAI-1 plays a regulatory role in tissue remodeling, cell migration, and intracellular signaling. Emerging evidence points to a role for PAI-1 in various pathological conditions, including cardiovascular diseases, cancer, and fibrosis. Targeting PAI-1 is therefore a promising therapeutic strategy in PAI-1-related pathologies. A class of small molecule inhibitors including TM5441 and TM5484, designed to bind the cleft in the central β-sheet A of PAI-1, showed to be potent PAI-1 inhibitors in vivo. However, their binding site has not yet been confirmed. Here, we report two X-ray crystallographic structures of PAI-1 in complex with TM5484. The structures revealed a binding site at the flexible joint region, which is distinct from the presumed binding site. Based on the structural analysis and biochemical data we propose a mechanism for the observed dose-dependent two-step mechanism of PAI-1 inhibition. By binding to the flexible joint region in PAI-1, TM5484 might restrict the structural flexibility of this region, thereby inducing a substrate form of PAI-1 followed by a conversion to an inert form.
Collapse
Affiliation(s)
- Machteld Sillen
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000 Leuven, Belgium;
| | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8577, Japan;
| | - Douglas E. Vaughan
- Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Sergei V. Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000 Leuven, Belgium;
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000 Leuven, Belgium;
- Correspondence:
| |
Collapse
|
103
|
Wang B, Sun W, Bi K, Li Y, Li F. Apremilast prevents IL‑17‑induced cellular senescence in ATDC5 chondrocytes mediated by SIRT1. Int J Mol Med 2021; 47:12. [PMID: 33448323 PMCID: PMC7834959 DOI: 10.3892/ijmm.2021.4845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/30/2020] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis is the most prevalent joint degenerative disease and has been considered a major cause of severe joint pain and physical disability in the elderly. The chondrocyte is the only cell type found in articular cartilage and chondrocyte senescence plays a pivotal role in the pathogenesis of osteoarthritis. Apremilast is an oral PDE4 inhibitor and has been used for the treatment of patients with active psoriatic arthritis. In the present study, the biological function of apremilast was examined in an interleukin (IL)-17-treated chondrocyte model. Expression levels of target genes and proteins were measured using reverse transcription-quantitative PCR, ELISA, and western blotting, respectively. ROS levels in chondrocytes were examined using the fluorescent dye DCFH-DA. Cellular senescence was determined using senescence-associated-β-galactosidase staining. The profile of cell cycle phases was analyzed via flow cytometry. It was revealed that treatment with apremilast reduced the expression of IL-1β, MCP-1, and the production of ROS. SA-β-gal staining results indicated that the presence of apremilast suppressed IL-17-induced cellular senescence. Furthermore, apremilast prevented IL-17-induced G0/G1 phase cell cycle arrest. In addition, it was demonstrated that apremilast suppressed IL-17-induced expression of p21 and PAI-1. Notably, the silencing of sirtuin 1 (SIRT1) abolished the protective effect of apremilast against IL-17-induced cellular senescence, suggesting that the action of apremilast in chondrocytes is dependent on SIRT1. In conclusion, the present results revealed that apremilast exerted a beneficial effect, thereby protecting chondrocytes from senescence induced by IL-17.
Collapse
Affiliation(s)
- Benlong Wang
- Department of Sports Medicine of Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, P.R. China
| | - Wei Sun
- Department of Sports Medicine of Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, P.R. China
| | - Kunwei Bi
- Department of Sports Medicine of Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, P.R. China
| | - Yong Li
- Department of Sports Medicine of Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, P.R. China
| | - Feng Li
- Department of Sports Medicine of Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277000, P.R. China
| |
Collapse
|
104
|
Liu R, Cheng F, Zeng K, Li W, Lan J. GPR120 Agonist GW9508 Ameliorated Cellular Senescence Induced by ox-LDL. ACS OMEGA 2020; 5:32195-32202. [PMID: 33376857 PMCID: PMC7758881 DOI: 10.1021/acsomega.0c03581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/15/2020] [Indexed: 05/03/2023]
Abstract
Introduction Oxidized low-density lipoprotein (ox-LDL)-induced endothelial senescence is involved in the pathogenesis of atherosclerosis and many cardiovascular diseases. G-protein-coupled receptor 120 (GPR120), a type of orphan G-protein-coupled receptors (GPRs), plays a vital role in mediating anti-inflammatory and insulin-sensitizing effects. The biological function of GPR120 in vascular endothelial cells is largely unknown. Methods The human aortic endothelial cells (HAECs) were treated with ox-LDL (100 μg/mL) in the presence or absence of GW9508 (50 μM) or AH9614 (1 μM) for 24 h. The LDH assay was used to determine cell death. The dihydroethidium (DHE) staining assay was used to measure intracellular levels of reactive oxidative species (ROS), and a senescence β-galactosidase assay kit was used to determine endothelial senescence. Gene and protein expressions were measured using real-time polymerase chain reaction (PCR) and western blot analysis, respectively. Results Ox-LDL treatment decreased the expression of GPR120 by more than half in HAECs. Typically, 100 μg/mL of ox-LDL- induced 35.2% LDH release, which was reduced to 16.9% by 50 μM GW9508, the agonist of GPR120. Importantly, GW9508 relieved cytotoxicity and suppressed the ox-LDL-induced increase in the activity of senescence-associated β-galactosidase (SA-β-Gal) (from 3.3-fold to 1.6-fold of the control group) and the generation of cellular reactive oxidative species (ROS) (from 3.8-fold to 1.6-fold of the control group). Furthermore, we found that GW9508 ameliorated ox-LDL-induced endothelial cell cycle arrest at the G0/G1 phase and the expression of key senescence proteins, including p53 and plasminogen activator inhibitor-1(PAI-1). Mechanistically, we showed that GW9508 promoted ox-LDL-induced transcriptional factor NF-E2-related factor 2 (NRF2) (increase by 47.3%) translocation into the nucleus. The effect of GW9508 is dependent on its receptor GPR120, the blockage of which by its specific antagonist, AH7614, abolished the antisenescence effect of GW9508. Conclusion Collectively, this study revealed the protective effect of GPR120 activation in vascular endothelial cells, implying that GPR120 is a promising therapeutic target for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Ruijie Liu
- Department
of Cardiology, The First Affiliated Hospital
of Ji’nan University, Guangzhou, Guangdong 510630, China
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
| | - Fei Cheng
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
| | - Kanghua Zeng
- Department
of Cardiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi 341000, China
| | - Wenfeng Li
- Department
of Cardiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi 341000, China
| | - Jun Lan
- Department
of Cardiology, Dongguan Songshanhu Central
Hospital, Dongguan, Guangdong 523326, China
- . Tel/Fax: +86-769-81368666
| |
Collapse
|
105
|
Sillen M, Declerck PJ. Targeting PAI-1 in Cardiovascular Disease: Structural Insights Into PAI-1 Functionality and Inhibition. Front Cardiovasc Med 2020; 7:622473. [PMID: 33415130 PMCID: PMC7782431 DOI: 10.3389/fcvm.2020.622473] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/03/2020] [Indexed: 01/31/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1), a member of the serine protease inhibitor (serpin) superfamily with antiprotease activity, is the main physiological inhibitor of tissue-type (tPA) and urokinase-type (uPA) plasminogen activators (PAs). Apart from being crucially involved in fibrinolysis and wound healing, PAI-1 plays a pivotal role in various acute and chronic pathophysiological processes, including cardiovascular disease, tissue fibrosis, cancer, and age-related diseases. In the prospect of treating the broad range of PAI-1-related pathologies, many efforts have been devoted to developing PAI-1 inhibitors. The use of these inhibitors, including low molecular weight molecules, peptides, antibodies, and antibody fragments, in various animal disease models has provided ample evidence of their beneficial effect in vivo and moved forward some of these inhibitors in clinical trials. However, none of these inhibitors is currently approved for therapeutic use in humans, mainly due to selectivity and toxicity issues. Furthermore, the conformational plasticity of PAI-1, which is unique among serpins, poses a real challenge in the identification and development of PAI-1 inhibitors. This review will provide an overview of the structural insights into PAI-1 functionality and modulation thereof and will highlight diverse approaches to inhibit PAI-1 activity.
Collapse
Affiliation(s)
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
106
|
Zhou H, Yan Y, Zhang X, Zhao T, Xu J, Han R. Ginseng polysaccharide inhibits MDA-MB-231 cell proliferation by activating the inflammatory response. Exp Ther Med 2020; 20:229. [PMID: 33149784 PMCID: PMC7604739 DOI: 10.3892/etm.2020.9359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Ginseng polysaccharide (GPS) is known for its efficacy in cancer therapy; however, its regulatory mechanism in breast cancer (BC) remains unclear. To analyze the effect of GPS on BC cell proliferation, cell proliferation rate calculations, western blotting, plasmid transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation assays were performed. GPS treatment in the culture cell medium inhibited cell proliferation in the BC cell line MDA-MB-231. In addition, the E-cadherin level was enhanced while the vimentin level was suppressed following GPS treatment (both P<0.05). Furthermore, the levels of apoptotic markers, including cleaved-Caspase-3 and p53, and inflammatory response markers, including plasminogen activator inhibitor and TNF-α, were induced by GPS treatment in MDA-MB-231 cells (all P<0.05). These results indicated that GPS supplementation activated the inflammatory response and apoptosis in BC cells. GPS treatment activated the phosphorylation levels of c-Jun N-terminal kinase, Akt and NF-κB. In MDA-MB-231 cells, GPS resulted in the accumulation of the NF-κB components p65, p50 and Ikaros family zing finger protein 1 (IKZF1; all, P<0.05). Chromatin immunoprecipitation and electrophoretic mobility shift assays indicated that p65 bound to the IKZF1 promoter. The overexpression of IKZF1 or p65 inhibited MDA-MB-231 cell proliferation (P<0.05), indicating that GPS treatment may inhibit BC cell proliferation by the activation of IKZF1. Taken together, these results suggested that GPS significantly inhibited BC cell proliferation via the control of the biological processes, including the activation of p65-IKZF1 signaling and apoptosis. The data indicated a novel mechanism for further understanding of cancer cell proliferation.
Collapse
Affiliation(s)
- Haoliang Zhou
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuxiang Yan
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Xianbo Zhang
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Ting Zhao
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Jiangang Xu
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Ruokuo Han
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
107
|
Sakurai S, Jojima T, Iijima T, Tomaru T, Usui I, Aso Y. Empagliflozin decreases the plasma concentration of plasminogen activator inhibitor-1 (PAI-1) in patients with type 2 diabetes: Association with improvement of fibrinolysis. J Diabetes Complications 2020; 34:107703. [PMID: 32883567 DOI: 10.1016/j.jdiacomp.2020.107703] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/21/2020] [Accepted: 07/26/2020] [Indexed: 12/30/2022]
Abstract
AIMS Elevation of the plasma concentration of plasminogen activator inhibitor-1 (PAI-1), a rapid-acting inhibitor of fibrinolysis, is associated with development of vascular thrombotic diseases, including coronary artery disease and stroke. We investigated the effects of empagliflozin, a sodium-glucose co-transporter-2 (SGLT2) inhibitor, on the plasma concentration of PAI-1 and fibrinolytic activity in patients with type 2 diabetes. METHODS In a randomized, active-controlled, open-label trial, 51 patients with type 2 diabetes were randomly allocated at a 2:1 ratio to receive empagliflozin (10 mg/day, n = 31) or standard therapy (n = 18) for 12 weeks. We measured the plasma concentrations of PAI-1 and plasmin-α2-antiplasmin complex (PAP) as indicators of fibrinolytic activity. Serum leptin and high-molecular weight (HMW) adiponectin were also measured. RESULTS In 49 patients who completed the trial, baseline plasma PAI-1 showed a positive correlation with body weight, visceral fat area (VFA), γ-glutamyltranspeptidase (GGT), leptin, and the platelet count, while it showed a negative correlation with HDL cholesterol and PAP. Body weight and VFA decreased significantly in the empagliflozin group, but not in the control group. The serum level of GGT showed a significant decrease at 12 weeks in the empagliflozin group, while it was unchanged in the control group. Serum HMW adiponectin increased significantly in the empagliflozin group. Plasma PAI-1 decreased significantly by 25% in the empagliflozin group, but not in the control group. In the empagliflozin group, the change of plasma PAI-1 was positively correlated with the changes of body weight and leptin, but was negatively correlated with the change of PAP. CONCLUSIONS Empagliflozin reduced the plasma PAI-1 concentration through its synergistic actions of a glucose-lowering effect, VFA loss, and restoring the adipokine balance. (Clinical trial registry: UMIN000025418).
Collapse
Affiliation(s)
- Shintaro Sakurai
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Teruo Jojima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan.
| | - Toshie Iijima
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Takuya Tomaru
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Yoshimasa Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi 321-0293, Japan.
| |
Collapse
|
108
|
Stress-induced RNA-chromatin interactions promote endothelial dysfunction. Nat Commun 2020; 11:5211. [PMID: 33060583 PMCID: PMC7566596 DOI: 10.1038/s41467-020-18957-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/11/2020] [Indexed: 01/06/2023] Open
Abstract
Chromatin-associated RNA (caRNA) has been proposed as a type of epigenomic modifier. Here, we test whether environmental stress can induce cellular dysfunction through modulating RNA-chromatin interactions. We induce endothelial cell (EC) dysfunction with high glucose and TNFα (H + T), that mimic the common stress in diabetes mellitus. We characterize the H + T-induced changes in gene expression by single cell (sc)RNA-seq, DNA interactions by Hi-C, and RNA-chromatin interactions by iMARGI. H + T induce inter-chromosomal RNA-chromatin interactions, particularly among the super enhancers. To test the causal relationship between H + T-induced RNA-chromatin interactions and the expression of EC dysfunction-related genes, we suppress the LINC00607 RNA. This suppression attenuates the expression of SERPINE1, a critical pro-inflammatory and pro-fibrotic gene. Furthermore, the changes of the co-expression gene network between diabetic and healthy donor-derived ECs corroborate the H + T-induced RNA-chromatin interactions. Taken together, caRNA-mediated dysregulation of gene expression modulates EC dysfunction, a crucial mechanism underlying numerous diseases. Global interaction of chromatin-associated RNAs and DNA can be identified in situ. Here the authors report the genome-wide increase of interchromosomal RNA-DNA interactions and demonstrate the importance of such RNA-DNA contacts exemplified by LINC00607 RNA and SERPINE1 gene’s super enhancer in dysfunctional endothelial cell models.
Collapse
|
109
|
Senescence-Associated Secretory Phenotype Determines Survival and Therapeutic Response in Cervical Cancer. Cancers (Basel) 2020; 12:cancers12102899. [PMID: 33050319 PMCID: PMC7601905 DOI: 10.3390/cancers12102899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 02/02/2023] Open
Abstract
Simple Summary Cervical cancer is the most common gynecological cancer caused by persistent infections with human papilloma viruses. Over time, this infection leads to secretion of inflammatory proteins in the cervix, which exacerbates the neoplastic and senescent changes to the cervical epithelial lining. We measured nineteen serum proteins in retrospectively collected samples from cervical cancer patients. We show here that 10 out of 19 proteins are associated with senescence phenotype in cervical cancer patients. This senescence associated protein signature influences how cervical cancer patients responds to therapy. Abstract Molecular biomarkers that can predict survival and therapeutic outcome are still lacking for cervical cancer. Here we measured a panel of 19 serum proteins in sera from 565 patients with stage II or III cervical cancer and identified 10 proteins that have an impact on disease specific survival (DSS) (Hazzard’s ratio; HR = 1.51–2.1). Surprisingly, all ten proteins are implicated in senescence-associated secreted phenotype (SASP), a hallmark of cellular senescence. Machine learning using Ridge regression of these SASP proteins can robustly stratify patients with high SASP, which is associated with poor survival, and patients with low SASP associated with good survival (HR = 3.09–4.52). Furthermore, brachytherapy, an effective therapy for cervical cancer, greatly improves survival in SASP-high patients (HR = 3.3, p < 5 × 10−5) but has little impact on survival of SASP-low patients (HR = 1.5, p = 0.31). These results demonstrate that cellular senescence is a major determining factor for survival and therapeutic response in cervical cancer and suggest that senescence reduction therapy may be an efficacious strategy to improve the therapeutic outcome of cervical cancer.
Collapse
|
110
|
Imerb N, Thonusin C, Chattipakorn N, Chattipakorn SC. Aging, obese-insulin resistance, and bone remodeling. Mech Ageing Dev 2020; 191:111335. [DOI: 10.1016/j.mad.2020.111335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 08/14/2020] [Indexed: 02/08/2023]
|
111
|
PAI-1, the Plasminogen System, and Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21197066. [PMID: 32993026 PMCID: PMC7582753 DOI: 10.3390/ijms21197066] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
The plasminogen system is a critical proteolytic system responsible for the remodeling of the extracellular matrix (ECM). The master regulator of the plasminogen system, plasminogen activator inhibitor-1 (PAI-1), has been implicated for its role in exacerbating various disease states not only through the accumulation of ECM (i.e., fibrosis) but also its role in altering cell fate/behaviour. Examination of PAI-1 has extended through various tissues and cell-types with recent investigations showing its presence in skeletal muscle. In skeletal muscle, the role of this protein has been implicated throughout the regeneration process, and in skeletal muscle pathologies (muscular dystrophy, diabetes, and aging-driven pathology). Needless to say, the complete function of this protein in skeletal muscle has yet to be fully elucidated. Given the importance of skeletal muscle in maintaining overall health and quality of life, it is critical to understand the alterations—particularly in PAI-1—that occur to negatively impact this organ. Thus, we provide a comprehensive review of the importance of PAI-1 in skeletal muscle health and function. We aim to shed light on the relevance of this protein in skeletal muscle and propose potential therapeutic approaches to aid in the maintenance of skeletal muscle health.
Collapse
|
112
|
Abstract
Within the past several decades, the emergence of new viral diseases with severe health complications and mortality is evidence of an age-dependent, compromised bodily response to abrupt stress with concomitantly reduced immunity. The new severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, causes coronavirus disease 2019 (COVID-19). It has increased morbidity and mortality in persons with underlying chronic diseases and those with a compromised immune system regardless of age and in older adults who are more likely to have these conditions. While SARS-CoV-2 is highly virulent, there is variability in the severity of the disease and its complications in humans. Severe pneumonia, acute respiratory distress syndrome, lung fibrosis, cardiovascular events, acute kidney injury, stroke, hospitalization, and mortality have been reported that result from pathogen-host interactions. Hallmarks of aging, interacting with one another, have been proposed to influence health span in older adults, possibly via mechanisms regulating the immune system. Here, we review the potential roles of the hallmarks of aging, coupled with host-coronavirus interactions. Of these hallmarks, we focused on those that directly or indirectly interact with viral infections, including immunosenescence, inflammation and inflammasomes, adaptive immunosenescence, genomic instability, mitochondrial dysfunction, epigenetic alterations, telomere attrition, and impaired autophagy. These hallmarks likely contribute to the increased pathophysiological responses to SARS-CoV-2 among older adults and may play roles as an additive risk of accelerated biological aging even after recovery. We also briefly discuss the role of antiaging drug candidates that require paramount attention in COVID-19 research.
Collapse
Affiliation(s)
- Shabnam Salimi
- Department of Epidemiology and Public Health, Division of Gerontology, University of Maryland School of Medicine, Baltimore
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
113
|
Bryant AG, Hu M, Carlyle BC, Arnold SE, Frosch MP, Das S, Hyman BT, Bennett RE. Cerebrovascular Senescence Is Associated With Tau Pathology in Alzheimer's Disease. Front Neurol 2020; 11:575953. [PMID: 33041998 PMCID: PMC7525127 DOI: 10.3389/fneur.2020.575953] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's Disease (AD) is associated with neuropathological changes, including aggregation of tau neurofibrillary tangles (NFTs) and amyloid-beta plaques. Mounting evidence indicates that vascular dysfunction also plays a key role in the pathogenesis and progression of AD, in part through endothelial dysfunction. Based on findings in animal models that tau pathology induces vascular abnormalities and cellular senescence, we hypothesized that tau pathology in the human AD brain leads to vascular senescence. To explore this hypothesis, we isolated intact microvessels from the dorsolateral prefrontal cortex (PFC, BA9) from 16 subjects with advanced Braak stages (Braak V/VI, B3) and 12 control subjects (Braak 0/I/II, B1), and quantified expression of 42 genes associated with senescence, cell adhesion, and various endothelial cell functions. Genes associated with endothelial senescence and leukocyte adhesion, including SERPINE1 (PAI-1), CXCL8 (IL8), CXCL1, CXCL2, ICAM-2, and TIE1, were significantly upregulated in B3 microvessels after adjusting for sex and cerebrovascular pathology. In particular, the senescence-associated secretory phenotype genes SERPINE1 and CXCL8 were upregulated by more than 2-fold in B3 microvessels after adjusting for sex, cerebrovascular pathology, and age at death. Protein quantification data from longitudinal plasma samples for a subset of 13 (n = 9 B3, n = 4 B1) subjects showed no significant differences in plasma senescence or adhesion-associated protein levels, suggesting that these changes were not associated with systemic vascular alterations. Future investigations of senescence biomarkers in both the peripheral and cortical vasculature could further elucidate links between tau pathology and vascular changes in human AD.
Collapse
Affiliation(s)
- Annie G Bryant
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Miwei Hu
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Becky C Carlyle
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Steven E Arnold
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Matthew P Frosch
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States.,Department of Pathology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Sudeshna Das
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Bradley T Hyman
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| | - Rachel E Bennett
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
114
|
Wei Z, Chang K, Fan C. Association between the 4G/5G polymorphism of plasminogen activator inhibitor-1 (PAI-1) gene and sudden sensorineural hearing loss in Caucasian population: a meta-analysis. Eur Arch Otorhinolaryngol 2020; 278:2203-2208. [PMID: 32901365 DOI: 10.1007/s00405-020-06305-z] [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: 03/23/2020] [Accepted: 08/18/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE To clarify the association between the 4G/5G polymorphism of plasminogen activator inhibitor-1 (PAI-1) and sudden sensorineural hearing loss (SSNHL). METHODS A systematic literature search of related studies up to August 30, 2019 in the PubMed and Embase databases was performed, and the results were displayed by odds ratios (ORs), and their 95% confidence intervals (CIs) were assessed using the STATA12.0 software using an allele model and a recessive model. RESULTS Three eligible studies covering 519 subjects (241 cases, 278 controls) were identified. No statistically significant association was detected between the 4G/5G polymorphism and SSNHL in any model (allele model: 5G vs. 4G, OR = 0.952, 95% CI = 0.765-1.185, P = 0.662; recessive model: 5G/5G vs. 4G/5G + 4G/4G, OR = 0.841, 95% CI = 0.415-1.704, P = 0.631). CONCLUSIONS There is no statistically significant association between the 4G/5G polymorphism of PAI-1 gene and SSNHL in the Caucasian population, and well-designed studies covering more patients and institutions should be conducted.
Collapse
Affiliation(s)
- Zhenxing Wei
- Department of Otorhinolaryngology-Head and Neck Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Middle No. 288 Zhongzhou Road, Luoyang, 471000, Henan, China.
| | - Kunpeng Chang
- Department of Otorhinolaryngology-Head and Neck Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Middle No. 288 Zhongzhou Road, Luoyang, 471000, Henan, China
| | - Chongsheng Fan
- Department of Otorhinolaryngology-Head and Neck Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Middle No. 288 Zhongzhou Road, Luoyang, 471000, Henan, China
| |
Collapse
|
115
|
Guo Y, Fan W, Xie Y, Cao S, Wan H, Jin B. SIRT1 Is the Target Gene for 2,3,5,4'-Tetrahydroxystilbene-2-O-β-D-Glucoside Alleviating the HUVEC Senescence. Front Pharmacol 2020; 11:542902. [PMID: 33013385 PMCID: PMC7508177 DOI: 10.3389/fphar.2020.542902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022] Open
Abstract
This study aimed to explore the effects of 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-d-glucoside (TSG) on the senescence of human umbilical vein cells (HUVEC) induced by hydrogen peroxide (H2O2) and to identify the potential targets mediating its protective action. HUVEC cells pre-treated with TSG for 24 h were exposed to H2O2 treatment. TSG significantly decreased H2O2-induced cellular senescence, as indicated by reduced senescence-associated β-galactosidase (SA-β-gal) positive staining, the proportion of cells in the G1 phase, cell apoptosis, p21, and plasminogen activator inhibitor-1 (PAI-1) expression. Moreover, TSG promoted Sirtuin 1 (SIRT1) expression. When SIRT1 was inhibited by EX527 or SIRT1 siRNA, the effect of TSG is diminished according to the increased proportion of cells in the G1 phase, cell apoptosis, p21, and PAI-1 expression. Overall, our study established TSG as an anti-senescence compound that exerts its protective action by regulating SIRT1 expression.
Collapse
Affiliation(s)
- Yan Guo
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China.,College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxue Fan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuefeng Xie
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyu Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
116
|
Maique J, Flores B, Shi M, Shepard S, Zhou Z, Yan S, Moe OW, Hu MC. High Phosphate Induces and Klotho Attenuates Kidney Epithelial Senescence and Fibrosis. Front Pharmacol 2020; 11:1273. [PMID: 32973510 PMCID: PMC7468469 DOI: 10.3389/fphar.2020.01273] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence is an irreversible cell growth arrest and is associated with aging and age-related diseases. High plasma phosphate (Pi) and deficiency of Klotho contribute to aging and kidney fibrosis, a pathological feature in the aging kidney and chronic kidney disease. This study examined the interactive role of Pi and Klotho in kidney senescence and fibrosis. Homozygous Klotho hypomorphic mice had high plasma Pi, undetectable Klotho in plasma and kidney, high senescence with massive collagen accumulation in kidney tubules, and fibrin deposits in peritubular capillaries. To examine the Pi effect on kidney senescence, a high (2%) Pi diet was given to wild-type mice. One week of high dietary Pi mildly increased plasma Pi, and upregulated kidney p16/p21 expression, but did not significantly decrease Klotho. Two weeks of high Pi intake led to increase in plasminogen activator inhibitor (PAI)-1, and decrease in kidney Klotho, but still without detectable increase in kidney fibrosis. More prolonged dietary Pi for 12 weeks exacerbated kidney senescence and fibrosis; more so in heterozygous Klotho hypomorphic mice compared to wild-type mice, and in mice with chronic kidney disease (CKD) on high Pi diet compared to CKD mice fed a normal Pi diet. In cultured kidney tubular cells, high Pi directly induced cellular senescence, injury and epithelial-mesenchymal transition, and enhanced H2O2-induced cellular senescence and injury, which were abrogated by Klotho. Fucoidan, a bioactive molecule with multiple biologic functions including senescence inhibition, blunted Pi-induced cellular senescence, oxidation, injury, epithelial-mesenchymal transition, and senescence-associated secretary phenotype. In conclusion, high Pi activates senescence through distinct but interconnected mechanisms: upregulating p16/p21 (early), and elevating plasminogen activator inhibitor-1 and downregulating Klotho (late). Klotho may be a promising agent to attenuate senescence and ameliorate age-associated, and Pi-induced kidney degeneration such as kidney fibrosis.
Collapse
Affiliation(s)
- Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brianna Flores
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sierra Shepard
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Zhiyong Zhou
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shirely Yan
- Departments of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
117
|
Khodamoradi E, Hoseini-Ghahfarokhi M, Amini P, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Targets for protection and mitigation of radiation injury. Cell Mol Life Sci 2020; 77:3129-3159. [PMID: 32072238 PMCID: PMC11104832 DOI: 10.1007/s00018-020-03479-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
Protection of normal tissues against toxic effects of ionizing radiation is a critical issue in clinical and environmental radiobiology. Investigations in recent decades have suggested potential targets that are involved in the protection against radiation-induced damages to normal tissues and can be proposed for mitigation of radiation injury. Emerging evidences have been shown to be in contrast to an old dogma in radiation biology; a major amount of reactive oxygen species (ROS) production and cell toxicity occur during some hours to years after exposure to ionizing radiation. This can be attributed to upregulation of inflammatory and fibrosis mediators, epigenetic changes and disruption of the normal metabolism of oxygen. In the current review, we explain the cellular and molecular changes following exposure of normal tissues to ionizing radiation. Furthermore, we review potential targets that can be proposed for protection and mitigation of radiation toxicity.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
- Misan Radiotherapy Center, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
118
|
Biomarkers of senescence in non-human primate adipose depots relate to aging. GeroScience 2020; 43:343-352. [PMID: 32705409 DOI: 10.1007/s11357-020-00230-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022] Open
Abstract
Forty-three female African green monkeys (Chlorocebus aethiops sabaeus) were selected to represent young adult to advanced geriatric ages (7-24 years) to exhibit a wide range of obesity status (8-53% body fat) and diverse metabolic syndrome criteria such as diabetes, dyslipidemia, and hypertension. Subcutaneous and visceral adipose tissues were collected and evaluated for the presence of senescence cells in both whole tissue and single-cell isolates from subcutaneous sources, utilizing senescence-associated β-galactosidase (SAβ-gal) staining. Plasma samples were analyzed for selected metabolic and inflammatory biomarkers related to the senescence-associated secretory profile. Our results indicated that tissue staining scores did not differ between subcutaneous and intra-abdominal visceral depots and were highly related within individuals. Tissue staining was significantly associated with chronological age; however, no associations with fatness or metabolic syndrome criteria were observed. Associations with age were unchanged when obesity status was included in regression models. Isolated cell staining did positively relate to age but not tissue staining, suggesting some of the SAβ-gal-positive cells were stromal vascular cells or small adipocytes, but that mature large adipocytes, filtered out in the cell isolation process, are also likely to exhibit positive SAβ-gal staining. Plasminogen activator inhibitor-1 (PAI-1) concentration in circulation was the sole inflammation-related biomarker that positively associated with age and is considered to be a marker of senescent cell burden. Our study is the largest, most comprehensive assessment of adipose SAβ-gal staining in a relevant animal model of human aging, and confirms that this senescence-associated biomarker specifically indicates an age-related process.
Collapse
|
119
|
Adnot S, Breau M, Houssaini A. PAI-1: A New Target for Controlling Lung-Cell Senescence and Fibrosis? Am J Respir Cell Mol Biol 2020; 62:271-272. [PMID: 31622556 PMCID: PMC7055697 DOI: 10.1165/rcmb.2019-0341ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Serge Adnot
- INSERM U955Créteil, France.,Département de Physiologie-Explorations Fonctionnelles, Hôpital Henri MondorCréteil, France.,Université Paris-Est CréteilCréteil, Franceand
| | - Marielle Breau
- Institut Paoli-Calmettes, Aix-Marseille UniversitéMarseille, France
| | | |
Collapse
|
120
|
Knockdown of angiopoietin-like 2 induces clearance of vascular endothelial senescent cells by apoptosis, promotes endothelial repair and slows atherogenesis in mice. Aging (Albany NY) 2020; 11:3832-3850. [PMID: 31186381 PMCID: PMC6594793 DOI: 10.18632/aging.102020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022]
Abstract
Elimination of senescent cells (SnC) is anti-atherogenic, but the specific contribution of senescent vascular endothelial cells (EC) is unknown. We inactivated angiopoietin like-2 (angptl2), a marker of SnEC and a pro-atherogenic cytokine in LDLr-/-, hApoB100+/+ atherosclerotic (ATX) mice. Three months after a single vascular delivery of a small hairpin (sh)Angptl2 in 3-month old ATX mice using an adeno-associated virus serotype 1 (AAV1), aortic atheroma plaque progression was slowed by 58% (p<0.0001). In the native aortic endothelium, angptl2 expression was decreased by 80%, in association with a reduced expression of p21, a cyclin-dependent kinase inhibitor overexpressed in growth-arrested SnC. Endothelial activation was reduced (lower Icam-1, Il-1β and Mcp-1 expression), decreasing monocyte Cd68 expression in the endothelium. One week post-injection, the ratio Bax/Bcl2 increased in the endothelium only, suggesting that angptl2+/p21+ SnEC were eliminated by apoptosis. Four weeks post-injection, the endothelial progenitor marker Cd34 increased, suggesting endothelial repair. In arteries of atherosclerotic patients, we observed a strong correlation between p21 and ANGPTL2 (r=0.727, p=0.0002) confirming the clinical significance of angptl2-associated senescence. Our data suggest that therapeutic down-regulation of vascular angptl2 leads to the clearance of SnEC by apoptosis, stimulates endothelial repair and reduces atherosclerosis.
Collapse
|
121
|
Roy-Vallejo E, Galván-Román JM, Moldenhauer F, Real de Asúa D. Adults with Down syndrome challenge another paradigm: When aging no longer entails arterial hypertension. J Clin Hypertens (Greenwich) 2020; 22:1127-1133. [PMID: 32644285 DOI: 10.1111/jch.13930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/09/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
The paradigmatic relationship between aging and atherosclerotic cardiovascular events does not apply to all patient populations. Though trisomy 21 (T21) and its phenotypic expression, Down syndrome (DS), are conditions that involve premature aging, the cardiovascular system of adults with DS appears to be particularly spared from this early senescence. Despite a higher prevalence of some classic cardiovascular risk factors in adults with DS than in the general population, such as dyslipidemia, obesity, or sedentarism, these individuals do not develop hypertension or suffer major cardiovascular events as they age. The protective factors that prevent the development of hypertension in T21 are not well established. Genes like RCAN1 and DYRK1A, both on chromosome 21 and over-expressed in adults with DS, appear to play a major role in cardiovascular prevention. Their regulation of the renin-angiotensin-aldosterone system (RAAS) and neprilysin synthesis could underlie the constitutive protection against arterial hypertension in adults with DS and explain the absence of increased arterial stiffness in this population. A better understanding of these molecular pathways could have enormous implications for the clinical management of adults with DS and might foster the development of novel therapeutic targets in cardiovascular prevention for the general population.
Collapse
Affiliation(s)
- Emilia Roy-Vallejo
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - José María Galván-Román
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Fernando Moldenhauer
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Diego Real de Asúa
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
122
|
Khoukaz HB, Ji Y, Braet DJ, Vadali M, Abdelhamid AA, Emal CD, Lawrence DA, Fay WP. Drug Targeting of Plasminogen Activator Inhibitor-1 Inhibits Metabolic Dysfunction and Atherosclerosis in a Murine Model of Metabolic Syndrome. Arterioscler Thromb Vasc Biol 2020; 40:1479-1490. [PMID: 32268785 PMCID: PMC7255962 DOI: 10.1161/atvbaha.119.313775] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Enhanced expression of PAI-1 (plasminogen activator inhibitor-1) has been implicated in atherosclerosis formation in humans with obesity and metabolic syndrome. However, little is known about the effects of pharmacological targeting of PAI-1 on atherogenesis. This study examined the effects of pharmacological PAI-1 inhibition on atherosclerosis formation in a murine model of obesity and metabolic syndrome. Approach and Results: LDL receptor-deficient (ldlr-/-) mice were fed a Western diet high in cholesterol, fat, and sucrose to induce obesity, metabolic dysfunction, and atherosclerosis. Western diet triggered significant upregulation of PAI-1 expression compared with normal diet controls. Addition of a pharmacological PAI-1 inhibitor (either PAI-039 or MDI-2268) to Western diet significantly inhibited obesity and atherosclerosis formation for up to 24 weeks without attenuating food consumption. Pharmacological PAI-1 inhibition significantly decreased macrophage accumulation and cell senescence in atherosclerotic plaques. Recombinant PAI-1 stimulated smooth muscle cell senescence, whereas a PAI-1 mutant defective in LRP1 (LDL receptor-related protein 1) binding did not. The prosenescent effect of PAI-1 was blocked by PAI-039 and R2629, a specific anti-LRP1 antibody. PAI-039 significantly decreased visceral adipose tissue inflammation, hyperglycemia, and hepatic triglyceride content without altering plasma lipid profiles. CONCLUSIONS Pharmacological targeting of PAI-1 inhibits atherosclerosis in mice with obesity and metabolic syndrome, while inhibiting macrophage accumulation and cell senescence in atherosclerotic plaques, as well as obesity-associated metabolic dysfunction. PAI-1 induces senescence of smooth muscle cells in an LRP1-dependent manner. These results help to define the role of PAI-1 in atherosclerosis formation and suggest a new plasma-lipid-independent strategy for inhibiting atherogenesis.
Collapse
Affiliation(s)
- Hekmat B Khoukaz
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Yan Ji
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Drew J Braet
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Manisha Vadali
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Ahmed A Abdelhamid
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti (C.D.E.)
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor (D.A.L.)
| | - William P Fay
- From the Department of Medicine (H.B.K, Y.J., D.J.B., M.V., A.A.A., W.P.F.), University of Missouri School of Medicine
- Department of Medical Pharmacology & Physiology (W.P.F.), University of Missouri School of Medicine
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (W.P.F.)
| |
Collapse
|
123
|
Chai B, Zheng ZH, Liao X, Li KY, Liang JS, Huang YX, Tong CJ, Ou DJ, Lu J. The protective role of omentin-1 in IL-1β-induced chondrocyte senescence. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:8-14. [PMID: 31852248 DOI: 10.1080/21691401.2019.1699803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Osteoarthritis is a common type of degenerative joint disease. Inflammation-related chondrocyte senescence plays a major role in the pathogenesis of osteoarthritis. Omentin-1 is a newly identified anti-inflammatory adipokine involved in lipid metabolism. In this study, we examined the biological function of omentin-1 in cultured chondrocytes. The presence of omentin-1 potently suppresses IL-1β-induced cellular senescence as revealed by staining with senescence-associated beta-galactosidase (SA-β-Gal). At the cellular level, omentin-1 attenuates IL-1β-induced G1 phase cell-cycle arrest. Mechanistically, we demonstrate that omentin-1 reduced IL-1β-induced expression of senescent factors including caveolin-1, p21, and PAI-1 as well as p53 acetylation through ameliorating SIRT1 reduction. Notably, silencing of SIRT1 abolishes IL-1β-induced senescence along with the induction of p21 and PAI-1, suggesting that the action of omentin-1 in chondrocytes is dependent on SIRT1. Collectively, our results revealed the molecular mechanism through which the adipokine omentin-1 exerts a beneficial effect, thereby protecting chondrocytes from senescence. Thus, omentin-1 could have clinical implication in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Bin Chai
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China.,Department of Orthopaedics, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, P. R. China
| | - Zi-Hui Zheng
- State Key Laboratory Cultivation Base For TCM Quality and Efficacy, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Xiang Liao
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Kang-Yang Li
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Jiang-Shan Liang
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Yong-Xiang Huang
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Chang-Jun Tong
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Di-Jun Ou
- Department of Orthopaedics, Shenzhen Nanshan People's Hospital, Shenzhen, P. R. China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P. R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P. R. China
| |
Collapse
|
124
|
Garcia V, Park EJ, Siragusa M, Frohlich F, Mahfuzul Haque M, Pascale JV, Heberlein KR, Isakson BE, Stuehr DJ, Sessa WC. Unbiased proteomics identifies plasminogen activator inhibitor-1 as a negative regulator of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:9497-9507. [PMID: 32300005 PMCID: PMC7196906 DOI: 10.1073/pnas.1918761117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.
Collapse
Affiliation(s)
- Victor Garcia
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Mauro Siragusa
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Florian Frohlich
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
- Department of Biology/Chemistry, Molecular Membrane Biology Section, University of Osnabrück, 49076 Osnabrück, Germany
| | - Mohammad Mahfuzul Haque
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Katherine R Heberlein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|
125
|
The Cbs Locus Affects the Expression of Senescence Markers and mtDNA Copy Number, but not Telomere Dynamics in Mice. Int J Mol Sci 2020; 21:ijms21072520. [PMID: 32260476 PMCID: PMC7177707 DOI: 10.3390/ijms21072520] [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: 01/04/2020] [Revised: 03/13/2020] [Accepted: 04/01/2020] [Indexed: 01/10/2023] Open
Abstract
Cystathionine β-synthase (CBS) is a housekeeping enzyme that catalyzes the first step of the homocysteine to cysteine transsulfuration pathway. Homozygous deletion of the Cbs gene in mice causes severe hyperhomocysteinemia and reduces life span. Here, we examined a possible involvement of senescence, mitochondrial DNA, and telomeres in the reduced life span of Cbs-/- mice. We found that senescence-related p21, Pai-1, Mcp1, and Il-6 mRNAs were significantly upregulated (2-10-fold) in liver, while p21 was upregulated in the brain of Cbs-/- mice (n = 20) compared with control Cbs+/- siblings (n = 20) in a sex- and age-dependent manner. Telomere length in blood (n = 80), liver (n = 40), and brain (n = 40) was not affected by the Cbs-/- genotype, but varied with sex and/or age. Levels of mitochondrial DNA tended to be reduced in livers, but not brains and blood, of Cbs-/- females (n = 20-40). The Cbs-/- genotype significantly reduced Tert mRNA expression in brain, but not liver, in a sex- and age-dependent manner. Multiple regression analysis showed that the senescence-related liver (but not brain) mRNAs and liver (but not brain or blood) mitochondrial DNA were associated with the Cbs genotype. In contrast, telomere length in blood, brain, and liver was not associated with the Cbs genotype or hyperhomocysteinemia, but was associated with sex (in brain and liver) and age (in brain and blood). Taken together, these findings suggest that the changes in senescence marker expression and mtDNA levels, but not telomere shortening, could account for the reduced life span of Cbs-/- mice.
Collapse
|
126
|
Dungan CM, Peck BD, Walton RG, Huang Z, Bamman MM, Kern PA, Peterson CA. In vivo analysis of γH2AX+ cells in skeletal muscle from aged and obese humans. FASEB J 2020; 34:7018-7035. [PMID: 32246795 DOI: 10.1096/fj.202000111rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022]
Abstract
Over the past 20 years, various identifiers of cellular senescence have been used to quantify the abundance of these cells in different tissues. These include classic markers such as p16, senescence-associated β-gal, and γH2AX, in addition to more recent markers (Sudan Black B and HMGB1). In vivo data on the usefulness of these markers in skeletal muscle are very limited and inconsistent. In the present study, we attempted to identify senescent cells in frozen human skeletal muscle biopsies using these markers to determine the effects of age and obesity on senescent cell burden; however, we were only able to assess the abundance of DNA-damaged nuclei using γH2AX immunohistochemistry. The abundance of γH2AX+ cells, including satellite cells, was not higher in muscle from old compared to young individuals; however, γH2AX+ cells were higher with obesity. Additionally, terminally differentiated, postmitotic myofiber nuclei from obese individuals had elevated γH2AX abundance compared to muscle from lean individuals. Analyses of gene expression support the conclusion that the elevated DNA damage and the senescence-associated secretory phenotype are preferentially associated with obesity in skeletal muscle. These data implicate obesity as a larger contributor to DNA damage in skeletal muscle than aging; however, more sensitive senescence markers for human skeletal muscle are needed to determine if these cells are in fact senescent.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Bailey D Peck
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - R Grace Walton
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Zhengyan Huang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Marcas M Bamman
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama, Birmingham, AL, USA.,Geriatric Research, Education, and Clinical Center, Birmingham VA Medical Center, Birmingham, AL, USA
| | - Philip A Kern
- Department of Internal Medicine/Endocrinology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Charlotte A Peterson
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
127
|
Jin CJ, Baumann A, Brandt A, Engstler AJ, Nier A, Hege M, Schmeer C, Kehm R, Höhn A, Grune T, Witte OW, Bergheim I. Aging-related liver degeneration is associated with increased bacterial endotoxin and lipopolysaccharide binding protein levels. Am J Physiol Gastrointest Liver Physiol 2020; 318:G736-G747. [PMID: 32090603 DOI: 10.1152/ajpgi.00345.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Aging is a risk factor in the development of many diseases, including liver-related diseases. The two aims of the present study were 1) to determine how aging affects liver health in mice in the absence of any interventions and 2) if degenerations observed in relation to blood endotoxin levels are critical in aging-associated liver degeneration. Endotoxin levels and markers of liver damage, mitochondrial dysfunction, insulin resistance, and apoptosis as well as the Toll-like receptor 4 (Tlr-4) signaling cascade were studied in liver tissue and blood, respectively, of 3- and 24-mo-old male C57BL/6J mice. In a second set of experiments, 3- to 4-mo-old and 14-mo-old female lipopolysaccharide-binding protein (LBP)-/- mice and littermates fed standard chow, markers of liver damage, insulin resistance, and mitochondrial dysfunction were assessed. Plasma activity of aspartate aminotransferase and histological signs of hepatic inflammation and fibrosis were significantly higher in old C57BL/6J mice than in young animals. The number of neutrophils, CD8α-positive cells, and mRNA expression of markers of apoptosis were also significantly higher in livers of old C57BL/6J mice compared with young animals, being also associated with a significant induction of hepatic Tlr-4 and LBP expression as well as higher endotoxin levels in peripheral blood. Compared with age-matched littermates, LBP-/- mice display less signs of senescence in liver. Taken together, our data suggest that, despite being fed standard chow, old mice developed liver inflammation and beginning fibrosis and that bacterial endotoxin may play a critical role herein.NEW & NOTEWORTHY Old age in mice is associated with marked signs of liver degeneration, hepatic inflammation, and fibrosis. Aging-associated liver degeneration is associated with elevated bacterial endotoxin levels and an induction of lipopolysaccharide-binding protein (LBP) and Toll-like receptor 4-dependent signaling cascades in liver tissue. Furthermore, in old aged LBP-/- mice, markers of senescence seem to be lessened, supporting the hypothesis that bacterial endotoxin levels might be critical in aging-associated decline of liver.
Collapse
Affiliation(s)
- Cheng Jun Jin
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Clinic for Gastroenterology, Hepatology, and Infectiology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anja Baumann
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Annette Brandt
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anna Janina Engstler
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Anika Nier
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| | - Marianne Hege
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christian Schmeer
- Hans-Berger Department of Neurology, University Hospital Jena, Jena, Germany
| | - Richard Kehm
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Annika Höhn
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Tilman Grune
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research, Munich-Neuherberg, Germany
| | - Otto W Witte
- Hans-Berger Department of Neurology, University Hospital Jena, Jena, Germany
| | - Ina Bergheim
- Institute of Nutrition, SD Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.,Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, Austria
| |
Collapse
|
128
|
Carmona P, Mendez N, Ili CG, Brebi P. The Role of Clock Genes in Fibrinolysis Regulation: Circadian Disturbance and Its Effect on Fibrinolytic Activity. Front Physiol 2020; 11:129. [PMID: 32231582 PMCID: PMC7083126 DOI: 10.3389/fphys.2020.00129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022] Open
Abstract
The fibrinolytic system is critical during the onset of fibrinolysis, a fundamental mechanism for fibrin degradation. Both tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) trigger fibrinolysis, leading to proteolytic activation of plasminogen to plasmin and subsequently fibrin proteolysis. This system is regulated by several inhibitors; plasminogen activator inhibitor-1 (PAI-1), the most studied, binds to and inactivates both tPA and uPA. Through the action of plasmin, this system regulates several physiological processes: embryogenesis, activation of inflammatory cells, cell proliferation and death, synaptic plasticity, wound healing, and others. The deregulated intervention of fibrinolysis in the pathophysiology of various diseases has been widely studied; findings of altered functioning have been reported in different chronic non-communicable diseases (NCD), reinforcing its pleiotropic character and the importance of its physiology and regulation. The evidence indicates that fundamental elements of the fibrinolytic system, such as tPA and PAI-1, show a circadian rhythm in their plasmatic levels and their gene expression are regulated by circadian system elements, known as clock genes – Bmal, Clock, Cry-, and accessory clock genes such as Rev-Erb and Ror. The disturbance in the molecular machinery of the clock by exposure to light during the night alters the natural light/dark cycle and causes disruption of the circadian rhythm. Such exposure affects the synchronization and functioning of peripheral clocks responsible for the expression of the components of the fibrinolytic system. So, this circadian disturbance could be critical in the pathophysiology of chronic diseases where this system has been found to be deregulated.
Collapse
Affiliation(s)
- Pamela Carmona
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Programa de Doctorado en Ciencias Médicas, Universidad de La Frontera, Temuco, Chile.,Laboratory of Integrative Biology, Center for Excellence in Translational Medicine, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Natalia Mendez
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Carmen G Ili
- Programa de Doctorado en Ciencias Médicas, Universidad de La Frontera, Temuco, Chile.,Laboratory of Integrative Biology, Center for Excellence in Translational Medicine, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Priscilla Brebi
- Programa de Doctorado en Ciencias Médicas, Universidad de La Frontera, Temuco, Chile.,Laboratory of Integrative Biology, Center for Excellence in Translational Medicine, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
129
|
Sillen M, Weeks SD, Zhou X, Komissarov AA, Florova G, Idell S, Strelkov SV, Declerck PJ. Molecular mechanism of two nanobodies that inhibit PAI-1 activity reveals a modulation at distinct stages of the PAI-1/plasminogen activator interaction. J Thromb Haemost 2020; 18:681-692. [PMID: 31858714 PMCID: PMC8855783 DOI: 10.1111/jth.14716] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1), a key inhibitor of plasminogen activators (PAs) tissue-type PA (tPA) and urokinase-type PA (uPA) plays a crucial role in many (patho)physiological processes (e.g., cardiovascular disease, tissue fibrosis) as well as in many age-related pathologies. Therefore, much effort has been put into the development of small molecule or antibody-based PAI-1 inhibitors. OBJECTIVE To elucidate the molecular mechanism of nanobody-induced PAI-1 inhibition. METHODS AND RESULTS Here we present the first crystal structures of PAI-1 in complex with two neutralizing nanobodies (Nbs). These structures, together with biochemical and biophysical characterization, reveal that Nb VHH-2g-42 (Nb42) interferes with the initial PAI-1/PA complex formation, whereas VHH-2w-64 (Nb64) redirects the PAI-1/PA interaction to PAI-1 deactivation and regeneration of active PA. Furthermore, whereas vitronectin does not have an impact on the inhibitory effect of Nb42, it strongly potentiates the inhibitory effect of Nb64, which may contribute to a strong inhibitory potential of Nb64 in vivo. CONCLUSIONS These findings illuminate the molecular mechanisms of PAI-1 inhibition. Nb42 and Nb64 can be used as starting points to engineer further improved antibody-based PAI-1 inhibitors or guide the rational design of small molecule inhibitors to treat a wide range of PAI-1-related pathophysiological conditions.
Collapse
Affiliation(s)
- Machteld Sillen
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Stephen D. Weeks
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Xiaohua Zhou
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Andrey A. Komissarov
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center, Tyler, TX, USA
| | - Galina Florova
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center, Tyler, TX, USA
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center, Tyler, TX, USA
| | - Sergei V. Strelkov
- Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
130
|
Rana T, Jiang C, Liu G, Miyata T, Antony V, Thannickal VJ, Liu RM. PAI-1 Regulation of TGF-β1-induced Alveolar Type II Cell Senescence, SASP Secretion, and SASP-mediated Activation of Alveolar Macrophages. Am J Respir Cell Mol Biol 2020; 62:319-330. [PMID: 31513752 PMCID: PMC7055702 DOI: 10.1165/rcmb.2019-0071oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022] Open
Abstract
Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-β1 (transforming growth factor-β1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-β1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-β1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-β1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.
Collapse
Affiliation(s)
- Tapasi Rana
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Chunsun Jiang
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gang Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University, Tohoku, Japan
| | - Veena Antony
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Victor J. Thannickal
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rui-Ming Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
131
|
Lowe DJ, Herzog M, Mosler T, Cohen H, Felton S, Beli P, Raj K, Galanty Y, Jackson SP. Chronic irradiation of human cells reduces histone levels and deregulates gene expression. Sci Rep 2020; 10:2200. [PMID: 32042076 PMCID: PMC7010678 DOI: 10.1038/s41598-020-59163-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/24/2020] [Indexed: 01/10/2023] Open
Abstract
Over the past decades, there have been huge advances in understanding cellular responses to ionising radiation (IR) and DNA damage. These studies, however, were mostly executed with cell lines and mice using single or multiple acute doses of radiation. Hence, relatively little is known about how continuous exposure to low dose ionising radiation affects normal cells and organisms, even though our cells are constantly exposed to low levels of radiation. We addressed this issue by examining the consequences of exposing human primary cells to continuous ionising γ-radiation delivered at 6-20 mGy/h. Although these dose rates are estimated to inflict fewer than a single DNA double-strand break (DSB) per hour per cell, they still caused dose-dependent reductions in cell proliferation and increased cellular senescence. We concomitantly observed histone protein levels to reduce by up to 40%, which in contrast to previous observations, was not mainly due to protein degradation but instead correlated with reduced histone gene expression. Histone reductions were accompanied by enlarged nuclear size paralleled by an increase in global transcription, including that of pro-inflammatory genes. Thus, chronic irradiation, even at low dose-rates, can induce cell senescence and alter gene expression via a hitherto uncharacterised epigenetic route. These features of chronic radiation represent a new aspect of radiation biology.
Collapse
Affiliation(s)
- Donna J Lowe
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, UK.
- Wellcome/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK.
| | - Mareike Herzog
- Wellcome/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK
| | | | - Howard Cohen
- Elizabeth House Surgery, Warlingham, Surrey, CR6 9LF, UK
| | - Sarah Felton
- Department of Dermatology, Churchill Hospital, Oxford, OX3 7LJ, UK
| | - Petra Beli
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Ken Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, UK
| | - Yaron Galanty
- Wellcome/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK.
| | - Stephen P Jackson
- Wellcome/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK.
| |
Collapse
|
132
|
Synergy between plasminogen activator inhibitor-1, α-synuclein, and neuroinflammation in Parkinson's disease. Med Hypotheses 2020; 138:109602. [PMID: 32035284 DOI: 10.1016/j.mehy.2020.109602] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a progressive degenerative nervous system disorder and is the second most common neurodegenerative disorder in the elderly population. The disease originates from the loss of dopamine-producing neurons in the substantia nigra in the brain, resulting in unregulated activity of the basal ganglia. Αlpha-synuclein (α-syn) is a protein found to aggregate in the substantia nigra region of patients with PD, forming Lewy Body inclusions; its aggregation may contribute to neuronal cell death in PD. This work hypothesizes about the synergistic relationship between α-syn aggregation and neuroinflammation to up-regulate expression of the serine protease inhibitor (serpin) plasminogen activator inhibitor-1 (PAI-1). The protease, plasmin, has been shown to cleave extracellular α-syn (including its monomeric, oligomeric, and fibrillary forms), resulting in less aggregation and Lewy Body formation. The zymogen plasminogen is converted to its active serine protease form, plasmin, either by tissue plasminogen activator (tPA) or by urokinase plasminogen activator (uPA) bound to urokinase receptor (uPAR). Both tPA and uPA/uPAR are inhibited by PAI-1. Thus, when PAI-1 levels increase, less plasmin is generated, which would lead to reduced proteolysis of α-syn. Expression of PAI-1 is increased both in inflammatory environments and in the presence of extracellular α-syn aggregates. This scenario suggests a pathological amplification loop: increased extracellular α-syn aggregation activates an inflammatory response from microglia and astrocytes, increasing PAI-1 levels, and decreasing the generation of plasmin. With reduced plasmin, less α-syn can be cleaved, and aggregation continues, sustaining the pathological process. Understanding this putative pathogenic loop could provide insight into the means by which neurodegeneration progresses in PD, and it may offer possible novel therapeutic strategies.
Collapse
|
133
|
Amiral J. Measurement of blood activation markers applied to the early diagnosis of cardiovascular alterations. Expert Rev Mol Diagn 2019; 20:85-98. [DOI: 10.1080/14737159.2020.1704258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jean Amiral
- Scientific-Hemostasis-Consulting, Scientific Director and Consultant in Thrombosis-Hemostasis, Andrésy, France
| |
Collapse
|
134
|
Candesartan Neuroprotection in Rat Primary Neurons Negatively Correlates with Aging and Senescence: a Transcriptomic Analysis. Mol Neurobiol 2019; 57:1656-1673. [PMID: 31811565 DOI: 10.1007/s12035-019-01800-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Preclinical experiments and clinical trials demonstrated that angiotensin II AT1 receptor overactivity associates with aging and cellular senescence and that AT1 receptor blockers (ARBs) protect from age-related brain disorders. In a primary neuronal culture submitted to glutamate excitotoxicity, gene set enrichment analysis (GSEA) revealed expression of several hundred genes altered by glutamate and normalized by candesartan correlated with changes in expression in Alzheimer's patient's hippocampus. To further establish whether our data correlated with gene expression alterations associated with aging and senescence, we compared our global transcriptional data with additional published datasets, including alterations in gene expression in the neocortex and cerebellum of old mice, human frontal cortex after age of 40, gene alterations in the Werner syndrome, rodent caloric restriction, Ras and oncogene-induced senescence in fibroblasts, and to tissues besides the brain such as the muscle and kidney. The most significant and enriched pathways associated with aging and senescence were positively correlated with alterations in gene expression in glutamate-injured neurons and, conversely, negatively correlated when the injured neurons were treated with candesartan. Our results involve multiple genes and pathways, including CAV1, CCND1, CDKN1A, CHEK1, ICAM1, IL-1B, IL-6, MAPK14, PTGS2, SERPINE1, and TP53, encoding proteins associated with aging and senescence hallmarks, such as inflammation, oxidative stress, cell cycle and mitochondrial function alterations, insulin resistance, genomic instability including telomere shortening and DNA damage, and the senescent-associated secretory phenotype. Our results demonstrate that AT1 receptor blockade ameliorates central mechanisms of aging and senescence. Using ARBs for prevention and treatment of age-related disorders has important translational value.
Collapse
|
135
|
Khalaf FA, Ibrahim HR, Bedair HM, Allam MM, Elshormilisy AA, Ali ST, Gaber WM. Plasminogen activator inhibitor-1 gene polymorphism as a risk factor for vascular complications in type 2 diabetes mellitus. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2019. [DOI: 10.1186/s43042-019-0018-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Diabetes mellitus (DM) can lead to microvascular and macrovascular damages through hyperglycemia that is the main cause of diabetic complications. Other factors such as hypertension, obesity, and hyperlipidemia may worsen or accelerate the others. Several studies have revealed definitive genetic predispositions to the development of type 2 diabetes mellitus (T2DM) and development of vascular complications. This study aimed to address the association between plasminogen activator inhibitor-1 (PAI-1) gene polymorphism and T2DM, and if this gene polymorphism may have a possible role in the development of vascular complications in T2DM. This study is a case control; it included 200 patients with T2DM, 117 patients had no vascular complications, and 83 had previous vascular complications (VCs). One hundred eighty volunteer blood donors were selected as a healthy control group. All patients and controls were subjected to clinical examination, and laboratory investigations included lipid profile, fasting and 2 h blood glucose, complete blood cell count, d-dimer, PAI-1, thrombin activatable fibrinolysis inhibitor (TAFI), and detection of PAI-1 gene polymorphism by real-time polymerase chain reaction (PCR).
Results
The most prevalent genotype of PAI-1 gene polymorphism in all studied groups, including controls, was 4G/5G with the highest allele frequency as 4G. The 4G/5G and 4G/4G genotypes were associated with increased risk of DM development as compared to 5G/5G genotype. The 4G/5G and 4G/4G genotypes also had a highly significant increased risk of VCs among diabetic patients, as compared to 5G/5G. The 4G allele also was highly associated with DM with VCs. The d-dimer TAFI, PAI-1 showed the highest levels in 4G/5G genotype followed by 4G/4G genotype. The lowest level was expressed in 5G/5G genotype in diabetic patients with and without VCs. The univariable analysis showed that genotypes 4G/5G and 4G/4G were potentially risk factors for development of VCs with T2DM patients.
Conclusion
This study concludes that the PAI-1 4G/5G polymorphism may be associated with T2DM and may be considered as a risk factor for development of thrombotic events. It may also help in selection and dosing of patients being treated with anticoagulant and fibrinolytic agents. Further large-scale studies are recommended to assess the possible role of environmental factors and gene interactions in the development of T2DM vascular risks.
Collapse
|
136
|
Vidal R, Wagner JUG, Braeuning C, Fischer C, Patrick R, Tombor L, Muhly-Reinholz M, John D, Kliem M, Conrad T, Guimarães-Camboa N, Harvey R, Dimmeler S, Sauer S. Transcriptional heterogeneity of fibroblasts is a hallmark of the aging heart. JCI Insight 2019; 4:131092. [PMID: 31723062 DOI: 10.1172/jci.insight.131092] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/02/2019] [Indexed: 12/27/2022] Open
Abstract
Aging is a major risk factor for cardiovascular disease. Although the impact of aging has been extensively studied, little is known regarding the aging processes in cells of the heart. Here we analyzed the transcriptomes of hearts of 12-week-old and 18-month-old mice by single-nucleus RNA-sequencing. Among all cell types, aged fibroblasts showed most significant differential gene expression, increased RNA dynamics, and network entropy. Aged fibroblasts exhibited significantly changed expression patterns of inflammatory, extracellular matrix organization angiogenesis, and osteogenic genes. Functional analyses indicated deterioration of paracrine signatures between fibroblasts and endothelial cells in old hearts. Aged heart-derived fibroblasts had impaired endothelial cell angiogenesis and autophagy and augmented proinflammatory response. In particular, expression of Serpine1 and Serpine2 were significantly increased and secreted by old fibroblasts to exert antiangiogenic effects on endothelial cells, an effect that could be significantly prevented by using neutralizing antibodies. Moreover, we found an enlarged subpopulation of aged fibroblasts expressing osteoblast genes in the epicardial layer associated with increased calcification. Taken together this study provides system-wide insights and identifies molecular changes of aging cardiac fibroblasts, which may contribute to declined heart function.
Collapse
Affiliation(s)
- Ramon Vidal
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Julian Uwe Gabriel Wagner
- Faculty for Biological Sciences and.,Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | - Ralph Patrick
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales Sydney, Kensington, New South Wales, Australia
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Marion Muhly-Reinholz
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - David John
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Magdalena Kliem
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Thomas Conrad
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nuno Guimarães-Camboa
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Richard Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, University of New South Wales Sydney, Kensington, New South Wales, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales Sydney, Kensington, New South Wales, Australia
| | - Stefanie Dimmeler
- Faculty for Biological Sciences and.,Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Centre for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
137
|
Griukova A, Deryabin P, Shatrova A, Burova E, Severino V, Farina A, Nikolsky N, Borodkina A. Molecular basis of senescence transmitting in the population of human endometrial stromal cells. Aging (Albany NY) 2019; 11:9912-9931. [PMID: 31689238 PMCID: PMC6874437 DOI: 10.18632/aging.102441] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Hormone-regulated proliferation and differentiation of endometrial stromal cells (ESCs) determine overall endometrial plasticity and receptivity to embryos. Previously we revealed that ESCs may undergo premature senescence, accompanied by proliferation loss and various intracellular alterations. Here we focused on whether and how senescence may be transmitted within the ESCs population. We revealed that senescent ESCs may induce paracrine senescence in young counterparts via cell contacts, secreted factors and extracellular vesicles. According to secretome-wide profiling we identified plasminogen activator inhibitor -1 (PAI-1) to be the most prominent protein secreted by senescent ESCs (data are available via ProteomeXchange with identifier PXD015742). By applying CRISPR/Cas9 techniques we disclosed that PAI-1 secreted by senescent ESCs may serve as the master-regulator of paracrine senescence progression within the ESCs population. Unraveled molecular basis of senescence transduction in the ESCs population may be further considered in terms of altered endometrial plasticity and sensitivity to invading embryo, thus contributing to the female infertility curing.
Collapse
Affiliation(s)
- Anastasiia Griukova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Pavel Deryabin
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Alla Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Elena Burova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Valeria Severino
- Department of Medicine, University Medical Center (CMU), Faculty of Medicine, Geneva University, Geneva CH-1211, Switzerland
| | - Annarita Farina
- Department of Medicine, University Medical Center (CMU), Faculty of Medicine, Geneva University, Geneva CH-1211, Switzerland
| | - Nikolay Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| | - Aleksandra Borodkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, Petersburg 194064, Russia
| |
Collapse
|
138
|
Jin G, Su Y, Dong Q, Zhao X, Zhang L, Yan X. Arctigenin alleviates TGF-β1-induced epithelial-mesenchymal transition and PAI-1 expression via AMPK/NF-κB pathway in peritoneal mesothelial cells. Biochem Biophys Res Commun 2019; 520:413-419. [PMID: 31607474 DOI: 10.1016/j.bbrc.2019.09.130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/27/2019] [Indexed: 12/24/2022]
Abstract
Peritoneal fibrosis (PF) caused by long-term peritoneal dialysis is closely associated with the epithelial-mesenchymal transition (EMT) of human peritoneal mesothelial cells (HPMCs). Moreover, the anti-fibrotic role of Arctigenin (Arc) has been reported in several fibrosis disorders. Therefore, the preventive effect of Arc on transforming growth factor-β1 (TGF-β1)-induced EMT and the underlying mechanisms in HPMCs was investigated in this study. Firstly, the PD model was established by TGF-β1 stimulation in cultured HPMCs in vitro, we found that TGF-β1 significantly increased the EMT markers (α-SMA, vimentin, and fibronectin) and plasminogen activator inhibitor type 1 (PAI-1) expressions, but decreased epithelial marker (E-cadherin). Co-treatment with Arc (10, 20, 40 μM) ameliorated TGF-β1-induced EMT in a dose-dependent manner, and the expression of PAI-1 was also inhibited by Arc, which was abrogated by restoration of PAI-1. Moreover, Arc enhanced the phosphorylated AMP-activated protein kinase (AMPK), but inhibited the phosphorylated IκBα level and nuclear translocation of nuclear factor κB (NF-κB) p65 in TGF-β1-induced HPMCs. ChIP and Luciferase reporter assays verified that the increased binding capacity of NF-κB to the promoter of PAI-1 induced by TGF-β1 was reversely attenuated by Arc in HPMCs. However, the effect of Arc on TGF-β1-induced NF-κB activation, PAI-1 expression and EMT in HPMCs was attenuated by AMPK agonist Compound C. In conclusion, these data demonstrated that Arc suppressed TGF-β1-induced EMT by activating the AMPK/NF-κB pathway to inhibit PAI-1 expression in HPMCs. Therefore, Arc might act as a potential therapeutic agent for PD treatment.
Collapse
Affiliation(s)
- Gang Jin
- Center of Kidney Dialysis, The Shaanxi Provincial People's Hospital, Xi'an, 710068, China; Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yanjin Su
- Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qianlan Dong
- Center of Kidney Dialysis, The Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Xiaohong Zhao
- Center of Kidney Dialysis, The Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Linping Zhang
- Center of Kidney Dialysis, The Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| | - Xiaohui Yan
- Center of Kidney Dialysis, The Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
139
|
TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int J Mol Sci 2019; 20:ijms20205002. [PMID: 31658594 PMCID: PMC6834140 DOI: 10.3390/ijms20205002] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022] Open
Abstract
Aging is broadly defined as the functional decline that occurs in all body systems. The accumulation of senescent cells is considered a hallmark of aging and thought to contribute to the aging pathologies. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates a myriad of cellular processes and has important roles in embryonic development, physiological tissue homeostasis, and various pathological conditions. TGF-β exerts potent growth inhibitory activities in various cell types, and multiple growth regulatory mechanisms have reportedly been linked to the phenotypes of cellular senescence and stem cell aging in previous studies. In addition, accumulated evidence has indicated a multifaceted association between TGF-β signaling and aging-associated disorders, including Alzheimer’s disease, muscle atrophy, and obesity. The findings regarding these diseases suggest that the impairment of TGF-β signaling in certain cell types and the upregulation of TGF-β ligands contribute to cell degeneration, tissue fibrosis, inflammation, decreased regeneration capacity, and metabolic malfunction. While the biological roles of TGF-β depend highly on cell types and cellular contexts, aging-associated changes are an important additional context which warrants further investigation to better understand the involvement in various diseases and develop therapeutic options. The present review summarizes the relationships between TGF-β signaling and cellular senescence, stem cell aging, and aging-related diseases.
Collapse
|
140
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
141
|
C 2-Ceramide-Induced Rb-Dominant Senescence-Like Phenotype Leads to Human Breast Cancer MCF-7 Escape from p53-Dependent Cell Death. Int J Mol Sci 2019; 20:ijms20174292. [PMID: 31480728 PMCID: PMC6747432 DOI: 10.3390/ijms20174292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/12/2019] [Accepted: 08/22/2019] [Indexed: 12/22/2022] Open
Abstract
Ceramide is a sphingolipid which regulates a variety of signaling pathways in eukaryotic cells. Exogenous ceramide has been shown to induce cellular apoptosis. In this study, we observed that exogenous ceramide induced two distinct morphologies of cell fate following C2-ceramide treatment between the two breast cancer cell lines MCF-7 (wild type p53) and MDA-MB-231 (mutant p53) cells. The growth assessment showed that C2-ceramide caused significant growth inhibition and apoptosis in MDA-MB-231 cells through down-regulating the expression of mutant p53 whereas up-regulating the expression of pro-apoptotic Bad, and the proteolytic activation of caspase-3. However, senescence-associated (SA)-β-galactosidase (β-gal) was regulated in MCF-7 cells after C2-ceramide treatment. The results of proliferation and apoptosis assays showed that MCF-7 cells were more resistant to C2-ceramide treatment compared to MDA-MB-231 cells. Furthermore, C2-ceramide treatment induced a time-responsive increase in Rb protein, a key regulator of senescence accompanied with the upregulation of both mRNA level and protein level of SA-genes PAI-1 and TGaseII in MCF-7 but not in MDA-MB-231 cells, suggesting that some cancer cells escape apoptosis through modulating senescence-like phenotype. The results of our present study depicted the mechanism of C2-ceramide-resistant breast cancer cells, which might benefit the strategic development of ceramide-based chemotherapeutics against cancer in the future.
Collapse
|
142
|
Sun T, Ghosh AK, Eren M, Miyata T, Vaughan DE. PAI-1 contributes to homocysteine-induced cellular senescence. Cell Signal 2019; 64:109394. [PMID: 31472244 DOI: 10.1016/j.cellsig.2019.109394] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/12/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022]
Abstract
Cellular Senescence is associated with organismal aging and related pathologies. Previously, we reported that plasminogen activator inhibitor-1 (PAI-1) is an essential mediator of senescence and a potential therapeutic target for preventing aging-related pathologies. In this study, we investigate the efficacies of PAI-1 inhibitors in both in vitro and in vivo models of homocysteine (Hcy)-induced cardiovascular aging. Elevated Hcy, a known risk factor of cardiovascular diseases, induces endothelial senescence as evidenced by increased senescence-associated β-Gal positivity (SA-β-Gal), flattened cellular morphology, and cylindrical appearance of cellular nuclei. Importantly, inhibition of PAI-1 by small molecule inhibitors reduces the number of SA-β-Gal positive cells, normalizes cellular morphology and nuclear shape. Furthermore, while Hcy induces the levels of senescence regulators PAI-1, p16, p53 and integrin β3, and suppresses catalase expression, treatment with PAI-1 inhibitors blocks the Hcy-induced stimulation of senescence cadres, and reverses the Hcy-induced suppression of catalase, indicating that PAI-1 specific small molecule inhibitors are efficient to prevent Hcy-induced cellular senescence. Our in vivo study shows that the levels of integrin β3, a recently identified potential regulator of cellular senescence, and its interaction with PAI-1 are significantly elevated in Hcy-treated heart tissues. In contrast, Hcy suppresses antioxidant gene regulator Nrf2 expression in hearts. However, co-treatment with PAI-1 inhibitor completely blocks the stimulation of Hcy-induced induction of integrin β3 and reverses Nrf2 expression. Collectively these in vitro and in vivo studies indicate that pharmacological inhibition of PAI-1 improves endothelial and cardiac health by suppressing the pro-senescence effects of hyperhomocysteinemia through suppression of Hcy-induced master regulators of cellular senescence PAI-1 and integrin β3. Therefore, PAI-1 inhibitors are promising drugs for amelioration of hyperhomocysteinemia-induced vascular aging and aging-related disease.
Collapse
Affiliation(s)
- Tianjiao Sun
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Toshio Miyata
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA; United Centers for Advanced Research and Translational Medicine, Tohoku University, Miyagi, Japan
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
143
|
Chan SL, Bishop N, Li Z, Cipolla MJ. Inhibition of PAI (Plasminogen Activator Inhibitor)-1 Improves Brain Collateral Perfusion and Injury After Acute Ischemic Stroke in Aged Hypertensive Rats. Stroke 2019; 49:1969-1976. [PMID: 29991657 DOI: 10.1161/strokeaha.118.022056] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background and Purpose- Aging and hypertension, comorbidities prevalent in the stroke population, are associated with poor collateral status and worsened stroke outcome. However, underlying mechanisms by which these conditions affect stroke outcome are not clear. We studied the role of PAI (plasminogen activator inhibitor)-1 that is increased in aging and hypertension on brain and vascular expression of inflammatory factors and perfusion that may contribute to worse stroke outcomes. Methods- Aged (≈50 weeks) and young (≈18 weeks) spontaneously hypertensive rats (SHR) were subjected to ischemia by middle cerebral artery occlusion (2 hours) and reperfusion (2 hours) with or without treatment with the PAI-1 inhibitor TM5441. Changes in middle cerebral artery and collateral perfusion territories were measured by multisite laser Doppler. Reactivity to TM5441 was studied using isolated and pressurized leptomeningeal anastomotic arterioles. Brain injury was determined by 2,3,5-triphenyltetrazolium staining and quantitative immunohistochemistry of amyloid-β-42, PAI-1, and hemoglobin. Circulating inflammatory factors were measured by ELISA. Results- Changes in cerebral blood flow during middle cerebral artery occlusion were similar between groups, with both having poor collateral perfusion and incomplete reperfusion. However, aged SHR had greater brain injury versus young (41±2 versus 23±2%, P<0.05) as well as increased brain deposition of amyloid-β-42 and circulating oxLDL (oxidized low-density lipoprotein). Erythrocyte aggregation and hemorrhage within the injured brain was observed in 50% of aged but no young SHR, with increased circulating PAI-1 in this subgroup of aged SHR (16±3 versus 6±2 ng/mL, P<0.05). PAI-1 inhibition with TM5441 improved brain injury but did not affect hemorrhage. TM5441 increased collateral perfusion by 38±7% and dilated leptomeningeal anastomotic arterioles by 44±10%, which was abolished by nitric oxide synthase inhibition. Conclusions- Increased injury in aged SHR seemed to be related to poor collateral perfusion, hemorrhagic transformation, increased amyloid-β-42, and oxidative stress. PAI-1 inhibition reduced infarction in both groups of SHR that possibly due, in part, to increased collateral perfusion.
Collapse
Affiliation(s)
- Siu-Lung Chan
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Nicole Bishop
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Zhaojin Li
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Marilyn J Cipolla
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.).,Obstetrics, Gynecology and Reproductive Sciences (M.J.C.).,Pharmacology (M.J.C.), University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
144
|
The TGF-β1/p53/PAI-1 Signaling Axis in Vascular Senescence: Role of Caveolin-1. Biomolecules 2019; 9:biom9080341. [PMID: 31382626 PMCID: PMC6723262 DOI: 10.3390/biom9080341] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022] Open
Abstract
Stress-induced premature cellular senescence is a significant factor in the onset of age-dependent disease in the cardiovascular system. Plasminogen activator inhibitor-1 (PAI-1), a major TGF-β1/p53 target gene and negative regulator of the plasmin-based pericellular proteolytic cascade, is elevated in arterial plaques, vessel fibrosis, arteriosclerosis, and thrombosis, correlating with increased tissue TGF-β1 levels. Additionally, PAI-1 is necessary and sufficient for the induction of p53-dependent replicative senescence. The mechanism of PAI-1 transcription in senescent cells appears to be dependent on caveolin-1 signaling. Src kinases are upstream effectors of both FAK and caveolin-1 activation as FAKY577,Y861 and caveolin-1Y14 phosphorylation are not detected in TGF-β1-stimulated src family kinase (pp60c-src, Yes, Fyn) triple-deficient (SYF−/−/−) cells. However, restoration of pp60c-src expression in SYF-null cells rescued both caveolin-1Y14 phosphorylation and PAI-1 induction in response to TGF-β1. Furthermore, TGF-β1-initiated Src phosphorylation of caveolin-1Y14 is critical in Rho-ROCK-mediated suppression of the SMAD phosphatase PPM1A maintaining and, accordingly, SMAD2/3-dependent transcription of the PAI-1 gene. Importantly, TGF-β1 failed to induce PAI-1 expression in caveolin-1-null cells, correlating with reductions in both Rho-GTP loading and SMAD2/3 phosphorylation. These findings implicate caveolin-1 in expression controls on specific TGF-β1/p53 responsive growth arrest genes. Indeed, up-regulation of caveolin-1 appears to stall cells in G0/G1 via activation of the p53/p21 cell cycle arrest pathway and restoration of caveolin-1 in caveolin-1-deficient cells rescues TGF-β1 inducibility of the PAI-1 gene. Although the mechanism is unclear, caveolin-1 inhibits p53/MDM2 complex formation resulting in p53 stabilization, induction of p53-target cell cycle arrest genes (including PAI-1), and entrance into premature senescence while stimulating the ATM→p53→p21 pathway. Identification of molecular events underlying senescence-associated PAI-1 expression in response to TGF-β1/src kinase/p53 signaling may provide novel targets for the therapy of cardiovascular disease.
Collapse
|
145
|
Wang M, Hao H, Leeper NJ, Zhu L. Thrombotic Regulation From the Endothelial Cell Perspectives. Arterioscler Thromb Vasc Biol 2019; 38:e90-e95. [PMID: 29793992 DOI: 10.1161/atvbaha.118.310367] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.) .,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Huifeng Hao
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.)
| | | | - Liyuan Zhu
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.)
| | | |
Collapse
|
146
|
Gustafson B, Nerstedt A, Smith U. Reduced subcutaneous adipogenesis in human hypertrophic obesity is linked to senescent precursor cells. Nat Commun 2019; 10:2757. [PMID: 31227697 PMCID: PMC6588633 DOI: 10.1038/s41467-019-10688-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/24/2019] [Indexed: 01/08/2023] Open
Abstract
Inappropriate expansion of the adipose cells in the subcutaneous adipose tissue (SAT) is a characteristic of hypertrophic obesity and of individuals with genetic predisposition for T2D (first-degree relatives; FDR). It is associated with insulin resistance, a dysfunctional, adipose tissue and reduced adipogenesis. We examined the regulation of adipogenesis in human SAT precursor cells and found ZNF521 to be a critical regulator of early adipogenic commitment and precursor cells leaving the cell cycle. However, neither altered upstream signalling nor lack of SAT progenitor cells could explain the reduced adipogenesis in hypertrophic obesity. Instead, we show that progenitor cells undergoing poor differentiation are characterized by senescence, inability to suppress p53/P16INK4 and secretion of factors reducing adipogenesis in non-senescent cells. We found aging, FDR and established T2D to be associated with increased progenitor cell senescence, reduced adipogenesis and hypertrophic expansion of the SAT adipose cells. Adipose tissue hypertrophy in obesity is associated with insulin resistance and other metabolic complications. Here, the authors analyze subcutaneous adipose tissue from patients with hypertrophic obesity and insulin resistance and find that adipocyte progenitor cells show features of senescence and have poor differentiation capacity.
Collapse
Affiliation(s)
- Birgit Gustafson
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, SE41345, Sweden
| | - Annika Nerstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, SE41345, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, SE41345, Sweden.
| |
Collapse
|
147
|
Delfarah A, Parrish S, Junge JA, Yang J, Seo F, Li S, Mac J, Wang P, Fraser SE, Graham NA. Inhibition of nucleotide synthesis promotes replicative senescence of human mammary epithelial cells. J Biol Chem 2019; 294:10564-10578. [PMID: 31138644 DOI: 10.1074/jbc.ra118.005806] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 05/18/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a mechanism by which cells permanently withdraw from the cell cycle in response to stresses including telomere shortening, DNA damage, or oncogenic signaling. Senescent cells contribute to both age-related degeneration and hyperplastic pathologies, including cancer. In culture, normal human epithelial cells enter senescence after a limited number of cell divisions, known as replicative senescence. Here, to investigate how metabolic pathways regulate replicative senescence, we used LC-MS-based metabolomics to analyze senescent primary human mammary epithelial cells (HMECs). We did not observe significant changes in glucose uptake or lactate secretion in senescent HMECs. However, analysis of intracellular metabolite pool sizes indicated that senescent cells exhibit depletion of metabolites from nucleotide synthesis pathways. Furthermore, stable isotope tracing with 13C-labeled glucose or glutamine revealed a dramatic blockage of flux of these two metabolites into nucleotide synthesis pathways in senescent HMECs. To test whether cellular immortalization would reverse these observations, we expressed telomerase in HMECs. In addition to preventing senescence, telomerase expression maintained metabolic flux from glucose into nucleotide synthesis pathways. Finally, we investigated whether inhibition of nucleotide synthesis in proliferating HMECs is sufficient to induce senescence. In proliferating HMECs, both pharmacological and genetic inhibition of ribonucleotide reductase regulatory subunit M2 (RRM2), a rate-limiting enzyme in dNTP synthesis, induced premature senescence with concomitantly decreased metabolic flux from glucose into nucleotide synthesis. Taken together, our results suggest that nucleotide synthesis inhibition plays a causative role in the establishment of replicative senescence in HMECs.
Collapse
Affiliation(s)
- Alireza Delfarah
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Sydney Parrish
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Jason A Junge
- the Translational Imaging Center, Molecular and Computational Biology, and
| | - Jesse Yang
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Frances Seo
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Si Li
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - John Mac
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Pin Wang
- From the Mork Family Department of Chemical Engineering and Materials Science
| | - Scott E Fraser
- the Translational Imaging Center, Molecular and Computational Biology, and
| | - Nicholas A Graham
- From the Mork Family Department of Chemical Engineering and Materials Science, .,the Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
148
|
Park HS, Kim ES, Ahn EH, Kim JO, An HJ, Kim JH, Lee Y, Lee WS, Kim YR, Kim NK. The microRNApolymorphisms inmiR-150 and miR-1179 are associated with risk of idiopathic recurrent pregnancy loss. Reprod Biomed Online 2019; 39:187-195. [PMID: 31182356 DOI: 10.1016/j.rbmo.2019.03.207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/22/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
RESEARCH QUESTION Are single nucleotide polymorphisms of microRNAs (miRNAs) and risk of idiopathic recurrent pregnancy loss (RPL) associated? DESIGN A total 375 patients with idiopathic RPL (age, mean ± standard deviation [SD] 33.02 ± 4.24 years; body mass index [BMI], mean ± SD, 21.57 ± 3.70 kg/m2) and 276 control participants (age, mean ± SD, 33.01 ± 5.27 years; BMI, mean ± SD, 21.58 ± 3.20) were recruited. Pregnancy loss was diagnosed using human chorionic gonadotrophin concentrations, ultrasonography and/or physical examination prior to 20 weeks of gestation. The genotype of the participants was determined by polymerase chain reaction restriction fragment length polymorphism analysis. Statistical analysis was performed to investigate the differences in frequencies between the control and RPL genotypes RESULTS: The miR-150G>A heterozygous genotype was significantly associated with increased risk of RPL (adjusted odds ratio 2.502, 95% confidence interval 1.555-4.025; P = 0.0002). The miR-1179A>T heterozygous genotype was significantly associated with decreased risk of RPL (adjusted odds ratio 0.633, 95% confidence interval 0.454-0.884; P = 0.007). Some allele combinations that included miR-150A or miRNA-1179T resulted in an increase or decrease in risk of RPL, respectively. CONCLUSIONS The miR-150G>A and miR-1179A>T polymorphisms were more frequently associated with RPL compared with controls.
Collapse
Affiliation(s)
- Han Sung Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea
| | - Eun Sun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea
| | - Eun Hee Ahn
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea
| | - Hui Jeong An
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Yubin Lee
- Fertility Center of CHA Gangnam Medical Center, CHA University Seoul, Republic of Korea
| | - Woo Sik Lee
- Fertility Center of CHA Gangnam Medical Center, CHA University Seoul, Republic of Korea
| | - Young Ran Kim
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
149
|
Yang SX, Zhang ZC, Bai HL. ClC-5 alleviates renal fibrosis in unilateral ureteral obstruction mice. Hum Cell 2019; 32:297-305. [PMID: 31054069 DOI: 10.1007/s13577-019-00253-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
Renal fibrosis is the major feature of end-stage renal disease with high mortality. Chloride (Cl-) moving along Cl- channels has been suggested to play to an important role in renal function. This study aims to investigate the role of ClC-5 in renal fibrosis in unilateral ureteral occlusion (UUO) mice. C57BL/6 mice received UUO surgery followed by delivery of adeno-associated virus encoding ClC-5 cDNA (AAVClC-5). Western blotting, real-time PCR and histological analysis were used to investigate the effects of ClC-5 on renal fibrosis and underlying mechanisms. The expression of ClC-5 was significantly decreased in renal cortex of UUO mice and transforming growth factor-β1 (TGF-β1)-stimulated HK2 cells. Overexpression of ClC-5 in vivo markedly ameliorated UUO-induced renal injury and fibrosis. The increased expressions of plasminogen activator inhibitor type 1, connective tissue growth factor, collagen III and collagen IV were also inhibited by ClC-5 upregulation. Moreover, UUO-induced immune cell infiltration and inflammatory cytokines release were attenuated in mice infected with AAVClC-5. In addition, the in vivo and in vitro results showed that ClC-5 overexpression prevented epithelial-to-mesenchymal transition (EMT), concomitantly with a restoration of E-cadherin expression and a decrease of vimentin, α-SMA and S100A4 expressions. Furthermore, ClC-5 overexpression inhibited UUO- or TGF-β1-induced increase in nuclear factor kappa B (NF-κB) acetylation and matrix metalloproteinases-9 (MMP-9) expression. However, downregulation of ClC-5 in HK2 cells further potentiated TGF-β1-induced EMT and increase in NF-κB acetylation and MMP-9 expression. ClC-5 upregulation ameliorates renal fibrosis via inhibiting NF-κB/MMP-9 pathway signaling activation, suggesting that ClC-5 may be a novel therapeutic target for treating renal fibrosis and chronic kidney disease.
Collapse
Affiliation(s)
- Shi-Xia Yang
- Department of Nephrology, Gansu Provincial Hospital of Traditional Chinese Medicine, No. 418 Guazhou Road, Lanzhou, 730050, Gansu, China.
| | - Zheng-Chang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hui-Ling Bai
- Department of Nephrology, Gansu Provincial Hospital of Traditional Chinese Medicine, No. 418 Guazhou Road, Lanzhou, 730050, Gansu, China
| |
Collapse
|
150
|
Shang L, Xue G, Gong L, Zhang Y, Peng S, Yuan C, Huang M. A novel ELISA for the detection of active form of plasminogen activator inhibitor-1 based on a highly specific trapping agent. Anal Chim Acta 2019; 1053:98-104. [DOI: 10.1016/j.aca.2018.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 11/26/2018] [Accepted: 12/08/2018] [Indexed: 10/27/2022]
|