1
|
Elmi M, Dass JH, Dass CR. The Various Roles of PEDF in Cancer. Cancers (Basel) 2024; 16:510. [PMID: 38339261 PMCID: PMC10854708 DOI: 10.3390/cancers16030510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a natural immunomodulator, anti-inflammatory, anti-angiogenic, anti-tumour growth and anti-metastasis factor, which can enhance tumour response to PEDF but can also conversely have pro-cancerous effects. Inflammation is a major cause of cancer, and it has been proven that PEDF has anti-inflammatory properties. PEDF's functional activity can be investigated through measuring metastatic and metabolic biomarkers that will be discussed in this review.
Collapse
Affiliation(s)
- Mitra Elmi
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| | - Joshua H. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Crispin R. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
2
|
Rai V, Le H, Agrawal DK. Novel mediators regulating angiogenesis in diabetic foot ulcer healing. Can J Physiol Pharmacol 2023; 101:488-501. [PMID: 37459652 DOI: 10.1139/cjpp-2023-0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
A non-healing diabetic foot ulcer (DFU) is a debilitating clinical problem amounting to socioeconomic and psychosocial burdens. DFUs increase morbidity due to prolonged treatment and mortality in the case of non-treatable ulcers resulting in gangrene and septicemia. The overall amputation rate of the lower extremity with DFU ranges from 3.34% to 42.83%. Wound debridement, antibiotics, applying growth factors, negative pressure wound therapy, hyperbaric oxygen therapy, topical oxygen, and skin grafts are common therapies for DFU. However, recurrence and nonhealing ulcers are still major issues. Chronicity of inflammation, hypoxic environment, poor angiogenesis, and decreased formation of the extracellular matrix (ECM) are common impediments leading to nonhealing patterns of DFUs. Angiogenesis is crucial for wound healing since proper vessel formation facilitates nutrients, oxygen, and immune cells to the ulcer tissue to help in clearing out debris and facilitate healing. However, poor angiogenesis due to decreased expression of angiogenic mediators and matrix formation results in nonhealing and ultimately amputation. Multiple proangiogenic mediators and vascular endothelial growth factor (VEGF) therapy exist to enhance angiogenesis, but the results are not satisfactory. Thus, there is a need to investigate novel pro-angiogenic mediators that can either alone or in combination enhance the angiogenesis and healing of DFUs. In this article, we critically reviewed the existing pro-angiogenic mediators followed by potentially novel factors that might play a regulatory role in promoting angiogenesis and wound healing in DFUs.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Hoangvi Le
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
3
|
Jones IC, Dass CR. Roles of pigment epithelium-derived factor in cardiomyocytes: implications for use as a cardioprotective therapeutic. J Pharm Pharmacol 2023:7146108. [PMID: 37104852 DOI: 10.1093/jpp/rgad037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES Cardiovascular diseases are the leading cause of death worldwide, with patients having limited options for treatment. Pigment epithelium-derived factor (PEDF) is an endogenous multifunctional protein with several mechanisms of action. Recently, PEDF has emerged as a potential cardioprotective agent in response to myocardial infarction. However, PEDF is also associated with pro-apoptotic effects, complicating its role in cardioprotection. This review summarises and compares knowledge of PEDF's activity in cardiomyocytes with other cell types and draws links between them. Following this, the review offers a novel perspective of PEDF's therapeutic potential and recommends future directions to understand the clinical potential of PEDF better. KEY FINDINGS PEDF's mechanisms as a pro-apoptotic and pro-survival protein are not well understood, despite PEDF's implication in several physiological and pathological activities. However, recent evidence suggests that PEDF may have significant cardioprotective properties mediated by key regulators dependent on cell type and context. CONCLUSIONS While PEDF's cardioprotective activity shares some key regulators with its apoptotic activity, cellular context and molecular features likely allow manipulation of PEDF's cellular activity, highlighting the importance of further investigation into its activities and its potential to be applied as a therapeutic to mitigate damage from a range of cardiac pathologies.
Collapse
Affiliation(s)
- Isobel C Jones
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| |
Collapse
|
4
|
Shao X, Hou X, Zhang X, Zhang R, Zhu R, Qi H, Zheng J, Guo X, Feng R. Integrated single-cell RNA-seq analysis reveals the vital cell types and dynamic development signature of atherosclerosis. Front Physiol 2023; 14:1118239. [PMID: 37089432 PMCID: PMC10117136 DOI: 10.3389/fphys.2023.1118239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/06/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: In the development of atherosclerosis, the remodeling of blood vessels is a key process involving plaque formation and rupture. So far, most reports mainly believe that macrophages, smooth muscle cells, and endothelial cells located at the intima and media of artery play the key role in this process. Few studies had focused on whether fibroblasts located at adventitia are involved in regulating disease process.Methods and results: In this study, we conducted in-depth analysis of single-cell RNA-seq data of the total of 18 samples from healthy and atherosclerotic arteries. This study combines several analysis methods including transcription regulator network, cell-cell communication network, pseudotime trajectory, gene set enrichment analysis, and differential expression analysis. We found that SERPINF1 is highly expressed in fibroblasts and is involved in the regulation of various signaling pathways.Conclusion: Our research reveals a potential mechanism of atherosclerosis, SERPINF1 regulates the formation and rupture of plaques through the Jak-STAT signaling pathway, which may provide new insights into the pathological study of disease. Moreover, we suggest that SRGN and IGKC as potential biomarkers for unstable arterial plaques.
Collapse
Affiliation(s)
- Xiuli Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiuyang Hou
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ruijia Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rongli Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - He Qi
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Jianling Zheng
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Xiaoling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
- *Correspondence: Rui Feng,
| |
Collapse
|
5
|
CircRNA-PTPRA Knockdown Inhibits Atherosclerosis Progression by Repressing ox-LDL-Induced Endothelial Cell Injury via Sponging of miR-671-5p. Biochem Genet 2023; 61:187-201. [PMID: 35817886 DOI: 10.1007/s10528-022-10256-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/22/2022] [Indexed: 01/24/2023]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease with high morbidity and mortality rates worldwide. This study aimed to investigate the role of circular RNA protein tyrosine phosphatase receptor type A (circRNA_PTPRA) in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cell (HUVECs) injury and its underlying molecular mechanism. The expression of circRNA-PTPRA and microRNA (miR)-671-5p was assessed by quantitative reverse transcription PCR (qRT-PCR). The interaction between circRNA-PTPRA and miR-671-5p was predicted using bioinformatic analysis. Cell viability and apoptosis were determined using the Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. Inflammation in HUVECs was analyzed by measuring the secretion of tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1β), and IL-6 using enzyme-linked immunosorbent assay (ELISA). Cleaved-caspase-3 expression was assessed using western blotting. The results indicated that circRNA-PTPRA expression was significantly increased and miR-671-5p expression was decreased in the serum of patients with AS and in ox-LDL-treated HUVECs. The interaction between circRNA-PTPRA and miR-671-5p was verified by dual luciferase reporter and RNA pull-down assays. In HUVECs, downregulation of circRNA-PTPRA reversed ox-LDL-induced reduction in cell viability, increase in apoptosis, and enhanced inflammation, whereas all these effects mediated by circRNA-PTPRA downregulation in ox-LDL-treated HUVECs were abolished by miR-671-5p downregulation. In conclusion, circRNA-PTPRA downregulation protects against ox-LDL-induced HUVECs injury by upregulating miR-671-5p, thereby providing potential therapeutic targets for AS.
Collapse
|
6
|
Sangaletti R, Tamames I, Yahn SL, Choi JS, Lee JK, King C, Rajguru SM. Mild therapeutic hypothermia protects against inflammatory and proapoptotic processes in the rat model of cochlear implant trauma. Hear Res 2023; 428:108680. [PMID: 36586170 PMCID: PMC9840707 DOI: 10.1016/j.heares.2022.108680] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Mild therapeutic hypothermia (MTH) has been demonstrated to prevent residual hearing loss from surgical trauma associated with cochlear implant (CI) insertion. Here, we aimed to characterize the mechanisms of MTH-induced hearing preservation in CI in a well-established preclinical rodent model. APPROACH Rats were divided into four experimental conditions: MTH-treated and implanted cochleae, cochleae implanted under normothermic conditions, MTH only cochleae and un-operated cochleae (controls). Auditory brainstem responses (ABRs) were recorded at different time points (up to 84 days) to confirm long-term protection and safety of MTH locally applied to the cochlea for 20 min before and after implantation. Transcriptome sequencing profiling was performed on cochleae harvested 24 h post CI and MTH treatment to investigate the potential beneficial effects and underlying active gene expression pathways targeted by the temperature management. RESULTS MTH treatment preserved residual hearing up to 3 months following CI when compared to the normothermic CI group. In addition, MTH applied locally to the cochleae using our surgical approach was safe and did not affect hearing in the long-term. Results of RNA sequencing analysis highlight positive modulation of signaling pathways and gene expression associated with an activation of cellular inflammatory and immune responses against the mechanical damage caused by electrode insertion. SIGNIFICANCE These data suggest that multiple and possibly independent molecular pathways play a role in the protection of residual hearing provided by MTH against the trauma of cochlear implantation.
Collapse
Affiliation(s)
- Rachele Sangaletti
- Department of Otolaryngology, University of Miami, Miami, FL, 33136, USA
| | - Ilmar Tamames
- Department of Biomedical Engineering, University of Miami, Miami, FL, 33136, USA
| | - Stephanie Lynn Yahn
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - James Seungyeon Choi
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | | | - Suhrud M Rajguru
- Department of Otolaryngology, University of Miami, Miami, FL, 33136, USA; Department of Biomedical Engineering, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
7
|
Szentimrei R, Lőrincz H, Szentpéteri A, E Varga V, Harangi M, Seres I, P Szabó R, Nemes B, Paragh G. Changes in serum pigment epithelium-derived factor levels after kidney transplantation in patients with end-stage renal disease. Ren Fail 2022; 44:1649-1659. [PMID: 36217673 PMCID: PMC9559055 DOI: 10.1080/0886022x.2022.2106243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background Pigment epithelium-derived factor (PEDF) is a serin protease inhibitor and a potent inhibitor of angiogenesis. Its serum level has significant associations with metabolic parameters. However, little is known about the association between PEDF levels and lipid parameters in renal transplanted (TX) patients. Therefore, our aim was to investigate the relationship between PEDF level and lipid parameters in TX patients. Methods Seventy TX patients (47 males, 23 females, mean age 51.7 ± 12.4 years) and 34 healthy controls were enrolled. We examined the serum creatinine, C-reactive protein, fasting glucose and lipid parameters right before, then 1 and 6 months after TX. High-density lipoprotein (HDL)-associated paraoxonase-1 (PON1) activities were measured spectrophotometrically. Lipoprotein subfractions were determined by Lipoprint. PEDF and oxidized low-density liporotein (oxLDL) levels were measured by ELISA. Results Before transplantation, patients had had a significantly higher PEDF level compared to control subjects (p < 0.001). One month after transplantation, their PEDF level decreased significantly reaching the healthy controls’ level, and this lower level was maintained during the 6 months follow-up period as well. The initial oxLDL level was significantly higher, while PON1 activities were significantly lower in the patient group compared to the control group. We found a significant positive correlation between PEDF and total cholesterol, low-density lipoprotein (LDL)-cholesterol, triglyceride, oxLDL and small HDL subfraction; while negative correlations were found between PEDF and mean LDL size and large HDL subfraction during the entire follow-up period. Conclusion PEDF may play an important role in the increased oxidative stress and enhanced atherogenesis in renal transplant patients.
Collapse
Affiliation(s)
- Réka Szentimrei
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Health Sciences, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Hajnalka Lőrincz
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Szentpéteri
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória E Varga
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mariann Harangi
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Seres
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Réka P Szabó
- Department of Nephrology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Nemes
- Department of Organ Transplantation, Faculty of Medicine, Institute of Surgery, University of Debrecen, Debrecen, Hungary
| | - György Paragh
- Division of Metabolic Disorders, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
8
|
Yang Y, Wang Z, Xu Y, Liu X, Sun Y, Li W. Knockdown of lncRNA H19 alleviates ox-LDL-induced HCAECs inflammation and injury by mediating miR-20a-5p/HDAC4 axis. Inflamm Res 2022; 71:1109-1121. [PMID: 35854140 DOI: 10.1007/s00011-022-01604-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: 11/15/2021] [Revised: 04/22/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) seriously disturbs the life of people. LncRNA H19 is reported to promote the progression of CAD; Nevertheless, the detailed mechanism by which H19 modulates CAD development is unclear. METHODS Clinical samples of CAD patients were collected, meanwhile we established in vitro and in vivo models of CAD by treating HCAECs with ox-LDL and feeding ApoE-/- mice with high fat diets (HFD). MTT assay was adopted to assess the cell viability. Transwell detection was applied to test the migration, and apoptosis was tested by flow cytometry. The levels of inflammatory cytokines were examined by ELISA. The relation among H19, miR-20a-5p and HDAC4 was explored by dual luciferase reporter and RIP assay. RESULTS H19 and HDAC4 levels were elevated, while miR-20a-5p was reduced in plasma of CAD patients and ox-LDL-treated HCAECs. ox-LDL increased H19 level and induced apoptosis and inflammation in HCAECs, while silencing of H19 rescued this phenomenon. In addition, the level of H19 was negatively correlated with miR-20a-5p, and miR-20a-5p inhibitor restored the effect of H19 silencing on HCAECs function. HDAC4 was the downstream mRNA of miR-20a-5p, and miR-20a-5p upregulation reversed ox-LDL-induced HCAECs injury through targeting HDAC4. Furthermore, H19 silencing significantly alleviated the coronary atherosclerotic plaques and inhibited the inflammatory responses in vivo. CONCLUSIONS We proved that knockdown of H19 alleviated ox-LDL-induced HCAECs injury via miR-20a-5p/HDAC4 axis, which might provide a new tactics against CAD.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Zhaofei Wang
- Department of Cardiology, Changsha First Hospital, Changsha, 410010, Hunan Province, People's Republic of China
| | - Ying Xu
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Xiaofang Liu
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Yehai Sun
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Wei Li
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China.
| |
Collapse
|
9
|
Wang Y, Liu X, Chao Z, Qin X, Quan X, Liu Z, Zhou Y, Jia C, Qin H, Zhang H. Pigment epithelium-derived factor maintains tight junction stability after myocardial infarction in rats through inhibition of the Wnt/β-catenin signaling pathway. Exp Cell Res 2022; 417:113213. [PMID: 35618012 DOI: 10.1016/j.yexcr.2022.113213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE The impairment of the coronary microcirculatory barrier caused by acute myocardial infarction (AMI) is closely related to poor prognosis. Recently, pigment epithelial-derived factor (PEDF) has been proven to be a promising cardiovascular protective drug. In this study, we demonstrated the protective role of PEDF in endothelial tight junctions (TJs) and the vascular barrier in AMI. MATERIALS AND METHODS 2, 3, 5-triphenyltetrazolium chloride (TTC), echocardiography and immunofluorescence staining were used to observe the size of infarcted myocardium area and cardiac function in myocardial tissue, and the distribution of tight junction proteins in human coronary endothelial cells (HCAEC). Dextran leakage assay and Transwell were used to assess the extent of vascular and HCAEC leakage. PCR and Western blot were used to detect tight junction-related mRNA and protein, and signaling pathway protein expression. RESULTS PEDF effectively reduced the infarction area and improved cardiac function in AMI rats, and lowered the leakage in AMI rats' angiocarpy and oxygen-glucose deprivation (OGD)-treated HCAEC. Furthermore, PEDF upregulated the expression of TJ mRNA and proteins in vivo and vitro. Mechanistically, PEDF inhibited the expression of phosphorylated low-density lipoprotein receptor-related protein 6 (p-LRP6) and active β-catenin under OGD, thus suppressing the activation of the classical Wnt pathway. CONCLUSIONS These novel findings demonstrated that PEDF maintained the expression of TJ proteins and endothelial barrier integrity by inhibiting the classical Wnt pathway during AMI.
Collapse
Affiliation(s)
- Yuzhuo Wang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Zhixiang Chao
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Xichun Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Xiaoyu Quan
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Zhiwei Liu
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yeqing Zhou
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Caili Jia
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Hao Qin
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Hao Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China; Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China.
| |
Collapse
|
10
|
Kanno Y, Shu E. α2-Antiplasmin as a Potential Therapeutic Target for Systemic Sclerosis. Life (Basel) 2022; 12:life12030396. [PMID: 35330147 PMCID: PMC8953682 DOI: 10.3390/life12030396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/24/2022] Open
Abstract
Systemic sclerosis is a connective tissue disease of unknown origin that is characterized by immune system abnormalities, vascular damage, and extensive fibrosis of the skin and visceral organs. α2-antiplasmin is known to be the main plasmin inhibitor and has various functions such as cell differentiation and cytokine production, as well as the regulation of the maintenance of the immune system, endothelial homeostasis, and extracellular matrix metabolism. The expression of α2-antiplasmin is elevated in dermal fibroblasts from systemic sclerosis patients, and the blockade of α2-antiplasmin suppresses fibrosis progression and vascular dysfunction in systemic sclerosis model mice. α2-antiplasmin may have promise as a potential therapeutic target for systemic sclerosis. This review considers the role of α2-antiplasmin in the progression of systemic sclerosis.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women’s College of Liberal Arts, 97-1 Kodo Kyotanabe, Kyoto 610-0395, Japan
- Department of Dermatology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan;
- Correspondence: ; Tel.:+81-0774-65-8629
| | - En Shu
- Department of Dermatology, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan;
| |
Collapse
|
11
|
He T, Liu W, Shen CA. Anti-inflammatory properties of pigment epithelium-derived factor. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221138857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inflammation is part of the complex biological response to harmful stimuli, such as cell damage, pathogens, or irritants. An excessive inflammatory response can lead to a variety of diseases. Pigment epithelium-derived factor (PEDF) is an endogenous glycoprotein that belongs to the superfamily of serine protease inhibitors and has multiple biological activities. Accumulating evidence suggests that PEDF participates in various inflammatory-related diseases, such as diabetic retinopathy, atherosclerosis, nonalcoholic steatohepatitis, and retinal diseases. However, the mechanism is still incompletely understood. In this paper, we review the anti-inflammatory properties of PEDF and discuss the underlying mechanisms. PEDF can exert its anti-inflammatory effects by downregulating the expression of inflammatory factors, promoting the synthesis of anti-inflammatory factors, inhibiting the activation of proinflammatory pathways and activating anti-inflammatory pathways. Examining the function of PEDF in inflammation addresses the need for further investigation and subsequent target-specific strategies for inflammatory disorders.
Collapse
Affiliation(s)
- Ting He
- The Fourth Medical Center of Chinese PLA General Hospital, Senior Department of Burns and Plastic Surgery, Beijing, China
| | - Wei Liu
- The Fourth Medical Center of Chinese PLA General Hospital, Senior Department of Burns and Plastic Surgery, Beijing, China
| | - Chuan-an Shen
- The Fourth Medical Center of Chinese PLA General Hospital, Senior Department of Burns and Plastic Surgery, Beijing, China
| |
Collapse
|
12
|
Yang X, Wang L, Zhang Z, Hu J, Liu X, Wen H, Liu M, Zhang X, Dai H, Ni M, Li R, Guo R, Zhang L, Luan X, Lin H, Dong M, Lu H. Ginsenoside Rb 1 Enhances Plaque Stability and Inhibits Adventitial Vasa Vasorum via the Modulation of miR-33 and PEDF. Front Cardiovasc Med 2021; 8:654670. [PMID: 34124194 PMCID: PMC8192703 DOI: 10.3389/fcvm.2021.654670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/20/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Atherosclerosis is closely associated with proliferation of the adventitial vasa vasorum, leading to the atherosclerotic plaque progression and vulnerability. In this report, we investigated the role of Ginsenoside Rb1 (Rb1) on atherosclerotic plaque stabilization and adventitial vasa vasorum (VV) along with the mechanisms involved. Methods and Results: Apolipoprotein E-deficient (ApoE-/-) mice were fed with a high-fat diet for 20 weeks, and then Ginsenoside Rb1 (50 mg/kg/d, intraperitoneal) was given for 4 weeks. Rb1 treatment significantly inhibited adventitial VV proliferation, alleviated inflammation, decreased plaque burden, and stabilized atherosclerotic plaques in apoE-/- mice. However, the beneficial effects of Rb1 on atherosclerotic lesion was attenuated by overexpression of miR-33. The analysis from atherosclerotic plaque revealed that Rb1 treatment could result in an induction of Pigment epithelium-derived factor (PEDF) expression and reduction of the miR-33 generation. Overexpression of miR-33 significantly reverted the Rb1-mediated elevation of PEDF and anti-angiogenic effect. Conclusions: Ginsenoside Rb1 attenuates plaque growth and enhances plaque stability partially through inhibiting adventitial vasa vasorum proliferation and inflammation in apoE-/- mice. The anti-angiogenic and anti-inflammation effects of Rb1 are exerted via the modulation of miR-33 and its target gene PEDF.
Collapse
Affiliation(s)
- Xiaoyan Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Lei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zihao Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiayi Hu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoling Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hao Wen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,The Second School of Clinical Medicine, Binzhou Medical University, Yantai, China
| | - Minghao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Hongyan Dai
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, China
| | - Mei Ni
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rui Li
- Department of Cardiology, China-Japan Friendship Hospital, Ministry of Health, Beijing, China
| | - Rong Guo
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Lei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaorong Luan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huili Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huixia Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
13
|
Ghadimi D, Nielsen A, Hassan MFY, Fölster-Holst R, Ebsen M, Frahm SO, Röcken C, de Vrese M, Heller KJ. Modulation of Proinflammatory Bacteria- and Lipid-Coupled Intracellular Signaling Pathways in a Transwell Triple Co-Culture Model by Commensal Bifidobacterium Animalis R101-8. Antiinflamm Antiallergy Agents Med Chem 2021; 20:161-181. [PMID: 33135616 DOI: 10.2174/1871523019999201029115618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Following a fat-rich diet, alterations in gut microbiota contribute to enhanced gut permeability, metabolic endotoxemia, and low grade inflammation-associated metabolic disorders. To better understand whether commensal bifidobacteria influence the expression of key metaflammation-related biomarkers (chemerin, MCP-1, PEDF) and modulate the pro-inflammatory bacteria- and lipid-coupled intracellular signaling pathways, we aimed at i) investigating the influence of the establishment of microbial signaling molecules-based cell-cell contacts on the involved intercellular communication between enterocytes, immune cells, and adipocytes, and ii) assessing their inflammatory mediators' expression profiles within an inflamed adipose tissue model. MATERIAL AND METHODS Bifidobacterium animalis R101-8 and Escherichia coli TG1, respectively, were added to the apical side of a triple co-culture model consisting of intestinal epithelial HT-29/B6 cell line, human monocyte-derived macrophage cells, and adipose-derived stem cell line in the absence or presence of LPS or palmitic acid. mRNA expression levels of key lipid metabolism genes HILPDA, MCP-1/CCL2, RARRES2, SCD, SFRP2 and TLR4 were determined using TaqMan qRT-PCR. Protein expression levels of cytokines (IL-1β, IL-6, and TNF-α), key metaflammation-related biomarkers including adipokines (chemerin and PEDF), chemokine (MCP- 1) as well as cellular triglycerides were assessed by cell-based ELISA, while those of p-ERK, p-JNK, p-p38, NF-κB, p-IκBα, pc-Fos, pc-Jun, and TLR4 were assessed by Western blotting. RESULTS B. animalis R101-8 inhibited LPS- and palmitic acid-induced protein expression of inflammatory cytokines IL-1β, IL-6, TNF-α concomitant with decreases in chemerin, MCP-1, PEDF, and cellular triglycerides, and blocked NF-kB and AP-1 activation pathway through inhibition of p- IκBα, pc-Jun, and pc-Fos phosphorylation. B. animalis R101-8 downregulated mRNA and protein levels of HILPDA, MCP-1/CCL2, RARRES2, SCD and SFRP2 and TLR4 following exposure to LPS and palmitic acid. CONCLUSION B. animalis R101-8 improves biomarkers of metaflammation through at least two molecular/signaling mechanisms triggered by pro-inflammatory bacteria/lipids. First, B. animalis R101-8 modulates the coupled intracellular signaling pathways via metabolizing saturated fatty acids and reducing available bioactive palmitic acid. Second, it inhibits NF-kB's and AP-1's transcriptional activities, resulting in the reduction of pro-inflammatory markers. Thus, the molecular basis may be formed by which commensal bifidobacteria improve intrinsic cellular tolerance against excess pro-inflammatory lipids and participate in homeostatic regulation of metabolic processes in vivo.
Collapse
Affiliation(s)
- Darab Ghadimi
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Annegret Nielsen
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | | | - Regina Fölster-Holst
- Clinic of Dermatology, University Hospital Schleswig-Holstein, Schittenhelmstr. 7, D-24105 Kiel, Germany
| | - Michael Ebsen
- Department of Pathology, Städtisches MVZ Kiel GmbH (Kiel City Hospital), Chemnitzstr.33, 24116 Kiel, Germany
| | - Sven Olaf Frahm
- Medizinisches Versorgungszentrum (MVZ), Pathology and Laboratory Medicine Dr. Rabenhorst, Prüner Gang 7, 24103 Kiel, Germany
| | - Christoph Röcken
- Institute of Pathology, Kiel University, University Hospital, Schleswig-Holstein, Arnold-Heller-Straße 3/14, D-24105 Kiel, Germany
| | - Michael de Vrese
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| | - Knut J Heller
- Department of Microbiology and Biotechnology, Max Rubner-Institute, Hermann-Weigmann-Str 1, D-24103 Kiel, Germany
| |
Collapse
|
14
|
Zou B, Huang T, Wu D, Hu X, Xiao L, Wang C, Zhang H, Xiang J, Hu C, Wu Q, Wu T. Knockdown of ZFAS1 improved the cardiac function of myocardial infarction rats via regulating Wnt/β-catenin signaling pathway. Aging (Albany NY) 2021; 13:12919-12928. [PMID: 33952724 PMCID: PMC8148456 DOI: 10.18632/aging.202961] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 03/14/2021] [Indexed: 01/21/2023]
Abstract
Myocardial infarction (MI) is a big health threat in the world, and it is characterized by high morbidity and mortality. However, current treatments are not effective enough, and novel therapeutic strategies need to be explored. ZFAS1 has been proved to be involved in the regulation of MI, but the specific mechanism remains unclear. MI rats were constructed through left anterior descending artery ligation, and hypoxia cell model was also established. The proliferation, invasion, and migration of cells were detected via CCK8, traswell, and wound healing methods. Immunohistochemistry staining, western blotting, and qRT-PCR were used to detect the levels of molecules. Knockdown of ZFAS1 significantly increased the proliferation, migration, and invasion of cardiac fibroblasts. Knockdown of ZFAS1 remarkably improved cardiac function via decreasing infarction ratio and increasing vWF expression, left ventricular ejection fraction, and left ventricular fractional shortening compared with group MI. Knockdown of ZFAS1 also suppressed Wnt/β-catenin pathway in vivo. The inhibition of Wnt/β-catenin remarkably reversed the influence of shZFAS1 on cardiac function and cardiac fibroblasts viability. Therefore, Knockdown of ZFAS1 could improve the cardiac function of myocardial infarction rats via regulating Wnt/β-catenin signaling pathway. The present study might provide new thoughts for the prevention and treatment of MI damage.
Collapse
Affiliation(s)
- Bing Zou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Dan Wu
- Jiangxi Health Vocational College, Nanchang, Jiangxi 330052, China
| | - Xiaomin Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Linghui Xiao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chenxi Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hongzhou Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Xiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Chenkai Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tao Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
15
|
Fujikawa T, Ohara M, Kohata Y, Nagaike H, Fukase A, Osaka N, Yashima H, Sato N, Kushima H, Shinmura K, Takahashi Y, Hiromura M, Terasaki M, Mori Y, Fukui T, Matsui T, Hirano T, Yamagishi SI. Glucose Variability is Independently Correlated with Serum Level of Pigment Epithelium-Derived Factor in Type 2 Diabetes. Diabetes Ther 2021; 12:827-842. [PMID: 33586119 PMCID: PMC7947132 DOI: 10.1007/s13300-021-01008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/20/2021] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Pigment epithelium-derived factor (PEDF) may play a role in cardiometabolic disorders. The aim of this study was to investigate which biochemical and clinical parameters are independently associated with serum PEDF levels in patients with type 2 diabetes mellitus (T2DM). METHODS We performed a cross-sectional analysis of 124 patients with T2DM who underwent continuous glucose monitoring (CGM) and blood chemistry analysis, including the diacron-reactive oxygen metabolites (d-ROMs) test and serum PEDF measurement (study 1). Then we investigated whether the changes in the studied biochemical and clinical parameters after 24 weeks of treatment (Δparameters) with anti-hyperglycemic agents, including sodium-glucose cotransporter 2 inhibitors, glucagon-like peptide 1 receptor agonists, and/or insulin and anti-hypertensive drugs and statins, were independently correlated with change in PEDF (ΔPEDF) in 52 of the patients with T2DM for whom there was sufficient serum samples to perform the post-treatment analysis (study 2). Serum levels of PEDF were measured with an enzyme-linked immunosorbent assay. CGM metrics were calculated on days 2 and 3. Oxidative stress was evaluated using the d-ROMs test. RESULTS Body mass index (BMI), triglycerides, fasting C-peptide, mean amplitude of glycemic excursions (MAGE), urinary albumin-to-creatinine ratio (UACR), and d-ROMs were positively associated with serum PEDF level, and high-density lipoprotein cholesterol (HDL-C) and estimated glomerular filtration rate (eGFR) were inversely associated with serum PEDF level. Because these parameters were correlated with each other, multivariate stepwise analysis was performed: eGFR, HDL-C, BMI, MAGE, and UACR remained significant (R2 = 0.452). Furthermore, ΔMAGE and Δd-ROMs were positively correlated with ΔPEDF in study 2. CONCLUSIONS The results of this study suggest that MAGE may be independently correlated with elevations in serum PEDF level in patients with T2DM.
Collapse
Affiliation(s)
- Tomoki Fujikawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Makoto Ohara
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Yo Kohata
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroe Nagaike
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ayako Fukase
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Naoya Osaka
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hironori Yashima
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Nobuko Sato
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideki Kushima
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kyoko Shinmura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yasuyoshi Takahashi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Munenori Hiromura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Michishige Terasaki
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yusaku Mori
- Anti-Glycation Research Section, Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Tomoyasu Fukui
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Diabetes Center, Ebina General Hospital, Ebina, Japan
| | - Sho-Ichi Yamagishi
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Ding Y, Feng Y, Zou Y, Wang F, Liu H, Liu C, Zhang Y. [Gly14]-humanin restores cathepsin D function via FPRL1 and promotes autophagic degradation of Ox-LDL in HUVECs. Nutr Metab Cardiovasc Dis 2020; 30:2406-2416. [PMID: 32917500 DOI: 10.1016/j.numecd.2020.07.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/01/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Abnormal aggregation of oxidized low-density lipoprotein (Ox-LDL) in vascular endothelial cells (VECs) is one of the major pathological changes in atherosclerotic lesions. Our research aimed to assess the mechanism of humanin (HN) in promoting autophagic degradation of Ox-LDL in HUVECs. METHODS AND RESULTS Flow cytometry and lipid quantitation results showed that Ox-LDL caused lipid and cholesterol accumulation in HUVECs. Western blot results showed that Ox-LDL increased the expression of autophagy-related proteins P62 and LC3-II in a concentration-dependent manner. The cathepsin D activity assay showed that Ox-LDL inhibited the function of cathepsin D. HNG pretreatment reduced lipid and cholesterol aggregation in HUVECs induced by Ox-LDL, increased LC3-II protein level, decreased P62 protein content, and reversed Ox-LDL-induced cathepsin D functional impairment. Inhibition of the FPRL1 pathway by FPRL1 siRNA or the FPRL1-specific inhibitor Boc-MLF blocked all HNG's protective effects. These results indicate that HNG could restore cathepsin D activity and protein level in HUVECs to repair lysosomal functional damage induced by Ox-LDL, further repairing Ox-LDL-induced autophagic damage in HUVECs. CONCLUSION HNG restores the activity of Ox-LDL-induced damaged lysosomal enzyme cathepsin D through its membrane protein receptor FPRL1 to promote autophagic degradation of Ox-LDL in HUVECs.
Collapse
Affiliation(s)
- Yu Ding
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yue Feng
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yutian Zou
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Fen Wang
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Huihui Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Chunfeng Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China; Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Yanlin Zhang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
17
|
Unung PJ, Bassey IE, Etukudo MH, Udoh AE, Alhassan MB, Akpan UO. Effect of glycemic control and dyslipidemia on plasma vascular endothelial growth factor and pigment epithelium-derived factor in diabetic retinopathy patients in Northern Nigeria. Int J Health Sci (Qassim) 2020; 14:4-12. [PMID: 33192226 PMCID: PMC7644452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES The disruption of the reciprocal regulation between vascular endothelial growth factor (VEGF) and pigment epithelium-derived factor (PEDF) has been associated with the pathogenesis of diabetic retinopathy (DR). This study assessed the levels of VEGF, PEDF, indices of glycemia, and lipid profile in diabetic patients with retinopathy. METHODS One hundred fifty participants comprised 50 type 2 diabetic patients with DR, 50 without DR and 50 non-diabetic normotensive controls, aged 30-80 years, were randomly recruited for this case-control study. The study was carried out from November 2017 to December 2018. VEGF, PEDF, glycated hemoglobin (HbA1c), fasting plasma glucose, and lipid profile were determined using standard methods. Blood pressures (BP) and anthropometric indices were measured. Chi-squared test of independence, analysis of variance, and Pearson's correlation were used to analyze data. Statistical significance was set at P < 0.05 and 95% confidence interval. RESULTS Both diabetic groups had significantly higher (P = 0.001) systolic and diastolic BP, VEGF, PEDF, HbA1c, fasting plasma glucose, triglycerides, total, high-density lipoprotein-cholesterol (HDL-C), and low-density lipoprotein-cholesterol (LDL-C) levels and significantly lower (P = 0.005) VEGF/PEDF than the controls. However, the diabetics with retinopathy had significantly higher (P = 0.001) HDL-C, LDL-C, VEGF, and PEDF levels compared to the diabetics without retinopathy. There were no significant differences (P > 0.05) in the levels of VEGF, PEDF, and VEGF/PEDF in both groups of diabetics that had good glycemic control and poor glycemic control. There was also no significant difference (P > 0.05) in the levels of VEGF and PEDF between the dyslipidemic and non-dyslipidemic subjects in both diabetic groups. CONCLUSION DR is associated with higher levels of VEGF and PEDF while good glycemic control and dyslipidemia seem not to have a profound effect on VEGF and PEDF levels in diabetics with or without DR. Higher PEDF levels are associated with higher atherogenic risk in the diabetics with retinopathy.
Collapse
Affiliation(s)
- Paulinus Jimmy Unung
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
- Department of Retinal Medicine, National Eye Centre, Kaduna, Nigeria
| | - Iya Eze Bassey
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Maisie Henrietta Etukudo
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Alphonsus Ekpe Udoh
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | | | - Uwem Okon Akpan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
18
|
Li S, Weinstein G, Zare H, Teumer A, Völker U, Friedrich N, Knol MJ, Satizabal CL, Petyuk VA, Adams HHH, Launer LJ, Bennett DA, De Jager PL, Grabe HJ, Ikram MA, Gudnason V, Yang Q, Seshadri S. The genetics of circulating BDNF: towards understanding the role of BDNF in brain structure and function in middle and old ages. Brain Commun 2020; 2:fcaa176. [PMID: 33345186 PMCID: PMC7734441 DOI: 10.1093/braincomms/fcaa176] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/04/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in brain development and function. Substantial amounts of BDNF are present in peripheral blood, and may serve as biomarkers for Alzheimer’s disease incidence as well as targets for intervention to reduce Alzheimer’s disease risk. With the exception of the genetic polymorphism in the BDNF gene, Val66Met, which has been extensively studied with regard to neurodegenerative diseases, the genetic variation that influences circulating BDNF levels is unknown. We aimed to explore the genetic determinants of circulating BDNF levels in order to clarify its mechanistic involvement in brain structure and function and Alzheimer’s disease pathophysiology in middle-aged and old adults. Thus, we conducted a meta-analysis of genome-wide association study of circulating BDNF in 11 785 middle- and old-aged individuals of European ancestry from the Age, Gene/Environment Susceptibility-Reykjavik Study (AGES), the Framingham Heart Study (FHS), the Rotterdam Study and the Study of Health in Pomerania (SHIP-Trend). Furthermore, we performed functional annotation analysis and related the genetic polymorphism influencing circulating BDNF to common Alzheimer’s disease pathologies from brain autopsies. Mendelian randomization was conducted to examine the possible causal role of circulating BDNF levels with various phenotypes including cognitive function, stroke, diabetes, cardiovascular disease, physical activity and diet patterns. Gene interaction networks analysis was also performed. The estimated heritability of BDNF levels was 30% (standard error = 0.0246, P-value = 4 × 10−48). We identified seven novel independent loci mapped near the BDNF gene and in BRD3, CSRNP1, KDELC2, RUNX1 (two single-nucleotide polymorphisms) and BDNF-AS. The expression of BDNF was associated with neurofibrillary tangles in brain tissues from the Religious Orders Study and Rush Memory and Aging Project (ROSMAP). Seven additional genes (ACAT1, ATM, NPAT, WDR48, TTC21A, SCN114 and COX7B) were identified through expression and protein quantitative trait loci analyses. Mendelian randomization analyses indicated a potential causal role of BDNF in cardioembolism. Lastly, Ingenuity Pathway Analysis placed circulating BDNF levels in four major networks. Our study provides novel insights into genes and molecular pathways associated with circulating BDNF levels and highlights the possible involvement of plaque instability as an underlying mechanism linking BDNF with brain neurodegeneration. These findings provide a foundation for a better understanding of BDNF regulation and function in the context of brain aging and neurodegenerative pathophysiology.
Collapse
Affiliation(s)
- Shuo Li
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Galit Weinstein
- School of Public Health, University of Haifa, Haifa 3498838, Israel
| | - Habil Zare
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA.,Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, 78229 TX, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Germany
| | - Maria J Knol
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, 78229 TX, USA.,Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX 78229, USA.,The Framingham Study, Framingham, MA 01702, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Hieab H H Adams
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, 3000 CA, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam 3015 CN, The Netherlands
| | - Lenore J Launer
- Department of Health and Human Services, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - David A Bennett
- Department of Neurology, Rush University Medical Center, Chicago, IL 60612, USA.,Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Philip L De Jager
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY 10032, USA.,Program in Population and Medical Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany.,German Center for Neurodegererative Diseases (DZNE), Rostock/Greifswald, Germany
| | - M Arfan Ikram
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Vilmundur Gudnason
- Faculty of Medicine, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland.,Icelandic Heart Association, 201 Kopavogur, Iceland
| | - Qiong Yang
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, 78229 TX, USA.,The Framingham Study, Framingham, MA 01702, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
19
|
Modulation of Lipid Metabolism by Trans-Anethole in Hepatocytes. Molecules 2020; 25:molecules25214946. [PMID: 33114589 PMCID: PMC7662808 DOI: 10.3390/molecules25214946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/12/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022] Open
Abstract
Non-alcoholic fatty liver disease is caused by excessive lipid accumulation in hepatocytes. Although trans-anethole (TAO) affects hypoglycemia and has anti-immune activity and anti-obesity effects, its role in non-alcoholic fatty liver disease remains unknown. This study aimed to evaluate the effects of TAO on cellular senescence, lipid metabolism, and reinforcement of microenvironments in HepG2 cells. To analyze the lipid metabolic activity of TAO, PCR analysis, flow-cytometry, and Oil Red O staining were performed, and mitochondrial membrane potential (MMP) and cellular senescence kits were used for assessing the suppression of cellular senescence. At 2000 μg/mL TAO, the cellular viability was approximately 99%, and cell senescence decreased dose-dependently. In the results for MMP, activity increased with concentration. The levels of lipolytic genes, CPT2, ACADS, and HSL, strongly increased over 3 days and the levels of lipogenic genes, ACC1 and GPAT, were downregulated on the first day at 1000 μg/mL TAO. Consequently, it was found that TAO affects the suppression of cellular senescence, activation of lipid metabolism, and reinforcement of the microenvironment in HepG2 cells, and can be added as a useful component to functional foods to prevent fatty liver disease and cellular senescence, as well as increase the immunoactivity of the liver.
Collapse
|
20
|
He D, Mao A, Zheng CB, Kan H, Zhang K, Zhang Z, Feng L, Ma X. Aortic heterogeneity across segments and under high fat/salt/glucose conditions at the single-cell level. Natl Sci Rev 2020; 7:881-896. [PMID: 34692110 PMCID: PMC8289085 DOI: 10.1093/nsr/nwaa038] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 12/24/2022] Open
Abstract
The aorta, with ascending, arch, thoracic and abdominal segments, responds to the heartbeat, senses metabolites and distributes blood to all parts of the body. However, the heterogeneity across aortic segments and how metabolic pathologies change it are not known. Here, a total of 216 612 individual cells from the ascending aorta, aortic arch, and thoracic and abdominal segments of mouse aortas under normal conditions or with high blood glucose levels, high dietary salt, or high fat intake were profiled using single-cell RNA sequencing. We generated a compendium of 10 distinct cell types, mainly endothelial (EC), smooth muscle (SMC), stromal and immune cells. The distributions of the different cells and their intercommunication were influenced by the hemodynamic microenvironment across anatomical segments, and the spatial heterogeneity of ECs and SMCs may contribute to differential vascular dilation and constriction that were measured by wire myography. Importantly, the composition of aortic cells, their gene expression profiles and their regulatory intercellular networks broadly changed in response to high fat/salt/glucose conditions. Notably, the abdominal aorta showed the most dramatic changes in cellular composition, particularly involving ECs, fibroblasts and myeloid cells with cardiovascular risk factor-related regulons and gene expression networks. Our study elucidates the nature and range of aortic cell diversity, with implications for the treatment of metabolic pathologies.
Collapse
Affiliation(s)
- Dongxu He
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Hao Kan
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhiming Zhang
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
21
|
de Lima AO, Koltes JE, Diniz WJS, de Oliveira PSN, Cesar ASM, Tizioto PC, Afonso J, de Souza MM, Petrini J, Rocha MIP, Cardoso TF, Neto AZ, Coutinho LL, Mourão GB, Regitano LCA. Potential Biomarkers for Feed Efficiency-Related Traits in Nelore Cattle Identified by Co-expression Network and Integrative Genomics Analyses. Front Genet 2020; 11:189. [PMID: 32194642 PMCID: PMC7064723 DOI: 10.3389/fgene.2020.00189] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Feed efficiency helps to reduce environmental impacts from livestock production, improving beef cattle profitability. We identified potential biomarkers (hub genes) for feed efficiency, by applying co-expression analysis in Longissimus thoracis RNA-Seq data from 180 Nelore steers. Six co-expression modules were associated with six feed efficiency-related traits (p-value ≤ 0.05). Within these modules, 391 hub genes were enriched for pathways as protein synthesis, muscle growth, and immune response. Trait-associated transcription factors (TFs) ELF1, ELK3, ETS1, FLI1, and TCF4, were identified with binding sites in at least one hub gene. Gene expression of CCDC80, FBLN5, SERPINF1, and OGN was associated with multiple feed efficiency-related traits (FDR ≤ 0.05) and were previously related to glucose homeostasis, oxidative stress, fat mass, and osteoblastogenesis, respectively. Potential regulatory elements were identified, integrating the hub genes with previous studies from our research group, such as the putative cis-regulatory elements (eQTLs) inferred as affecting the PCDH18 and SPARCL1 hub genes related to immune system and adipogenesis, respectively. Therefore, our analyses contribute to a better understanding of the biological mechanisms underlying feed efficiency in bovine and the hub genes disclosed can be used as biomarkers for feed efficiency-related traits in Nelore cattle.
Collapse
Affiliation(s)
- Andressa O de Lima
- Center for Biological and Health Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - James E Koltes
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Wellison J S Diniz
- Center for Biological and Health Sciences, Federal University of São Carlos, São Carlos, Brazil
| | | | - Aline S M Cesar
- Department of Agroindustry, Food and Nutrition, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, Brazil
| | | | - Juliana Afonso
- Center for Biological and Health Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Marcela M de Souza
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Juliana Petrini
- Exact Sciences Institute, Federal University of Alfenas, Alfenas, Brazil
| | - Marina I P Rocha
- Center for Biological and Health Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Tainã F Cardoso
- Embrapa Pecuária Sudeste, Empresa Brazileira de Pesquisa Agropecuária, São Carlos, Brazil
| | - Adhemar Zerlotini Neto
- Embrapa Informática Agropecuária, Empresa Brazileira de Pesquisa Agropecuária, Campinas, Brazil
| | - Luiz L Coutinho
- Department of Animal Science, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, Brazil
| | - Gerson B Mourão
- Department of Animal Science, Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, Brazil
| | - Luciana C A Regitano
- Embrapa Pecuária Sudeste, Empresa Brazileira de Pesquisa Agropecuária, São Carlos, Brazil
| |
Collapse
|
22
|
Fan Z, Zhang Y, Xiao D, Ma J, Liu H, Shen L, Zhang M, He B. Long noncoding RNA UC.98 stabilizes atherosclerotic plaques by promoting the proliferation and adhesive capacity in murine aortic endothelial cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:141-149. [PMID: 31942916 DOI: 10.1093/abbs/gmz155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/25/2022] Open
Abstract
Pathological studies have shown that the vulnerability of plaques affects outcomes in patients with atherosclerosis (AS), a chronic inflammatory disease and common cause of morbidity and mortality worldwide. Although emerging technologies have enabled early diagnosis of AS with high-risk vulnerable plaques, more accurate and noninvasive diagnostic methods are urgently required. To this end, molecules involved in genetic or epigenetic regulation of the vulnerability of atherosclerotic plaques have been extensively studied. Here, we evaluated long noncoding RNA (lncRNA) variability by microarray assay in murine aortic endothelial cells (MAECs) bearing vulnerable plaques and identified the novel functional lncRNA UC.98, whose expression pattern was associated with the vulnerability of atherosclerotic plaques. Consistent with this, clinical statistics comparing the peripheral blood specimens from sets of patients with AS with or without vulnerable plaques confirmed the linear relationship between the expression pattern of UC.98 and plaque instability. Moreover, MTT assays and western blot analysis showed that silencing of intrinsic UC.98 in MAECs not only suppressed cell proliferation but also decreased the expressions of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, thereby inactivating the nuclear factor-κB pathway. In conclusion, our results highlighted the pivotal role of UC.98 in regulating the vulnerability of plaques during AS progression and suggested that UC.98 may be a biomarker of the early diagnosis and prognosis of AS with vulnerable plaques and a potential therapeutic target for slowing AS progression.
Collapse
Affiliation(s)
- Zixu Fan
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ying Zhang
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Danrui Xiao
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Jianwei Ma
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Hua Liu
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Linghong Shen
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Min Zhang
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| | - Ben He
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China
| |
Collapse
|
23
|
Brook N, Brook E, Dharmarajan A, Chan A, Dass CR. The role of pigment epithelium-derived factor in protecting against cellular stress. Free Radic Res 2019; 53:1166-1180. [PMID: 31760841 DOI: 10.1080/10715762.2019.1697809] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Since its discovery as a neurotrophic factor in retinal pigmented epithelium cells in the late 1980s, there has been an increase in understanding of the role that pigment epithelium-derived factor (PEDF) plays in cellular functions. PEDF plays an important role in mediating cellular protection during exposure to oxidative stress and inflammation by preventing stress-induced angiogenesis and apoptosis. PEDF acts to reduce oxidative stress by promoting mitochondrial stability and by regulating the expression of enzymes involved in ROS accumulation and clearance. PEDF protects against the negative effects of oxidative stress by regulating cell survival pathways and the expression of inflammatory and proangiogenic mediators. PEDF-mediated cellular protection may be of clinical importance in diseases characterised by oxidative stress, chronic inflammation and pathological neovascularization, indicating that targeting PEDF may be a potential focus for therapeutic interventions in chronic diseases. In this review, we provide a historical perspective on the discoveries of PEDF interactions and functions, and discuss recent in vitro, in vivo and clinical findings to provide a current summary of the important protective effects following cellular exposure to stress stimuli and future clinical potential of PEDF.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Arun Dharmarajan
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia.,Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley, Australia.,Hollywood Private Hospital, Breast Clinical Trials Unit, Breast Cancer Research Centre-Western Australia, Nedlands, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|