1
|
Zhu Z, Li L, Ye Y, Zhong Q. Integrating bulk and single-cell transcriptomics to elucidate the role and potential mechanisms of autophagy in aging tissue. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00996-w. [PMID: 39414741 DOI: 10.1007/s13402-024-00996-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 10/18/2024] Open
Abstract
PURPOSE Autophagy is frequently observed in tissues during the aging process, yet the tissues most strongly correlated with autophagy during aging and the underlying regulatory mechanisms remain inadequately understood. The purpose of this study is to identify the tissues with the highest correlation between autophagy and aging, and to explore the functions and mechanisms of autophagy in the aging tissue microenvironment. METHODS Integrated bulk RNA-seq from over 7000 normal tissue samples, single-cell sequencing data from blood samples of different ages, more than 2000 acute myeloid leukemia (AML) bulk RNA-seq, and multiple sets of AML single-cell data. The datasets were analysed using various bioinformatic approaches. RESULTS Blood tissue exhibited the highest positive correlation between autophagy and aging among healthy tissues. Single-cell resolution analysis revealed that in aged blood, classical monocytes (C. monocytes) are most closely associated with elevated autophagy levels. Increased autophagy in these monocytes correlated with a higher proportion of C. monocytes, with hypoxia identified as a crucial contributing factor. In AML, a representative myeloid blood disease, enhanced autophagy was accompanied by an increased proportionof C. monocytes. High autophagy levels in monocytes are associated with pro-inflammatory gene upregulation and Reactive Oxygen Species (ROS) accumulation, contributing to tissue aging. CONCLUSION This study revealed that autophagy is most strongly correlated with aging in blood tissue. Enhanced autophagy levels in C. monocytes demonstrate a positive correlation with increased secretion of pro-inflammatory factors and elevated production of ROS, which may contribute to a more rapid aging process. This discovery underscores the critical role of autophagy in blood aging and suggests potential therapeutic targets to mitigate aging-related health issues.
Collapse
Affiliation(s)
- Zhenhua Zhu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linsen Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Liu S, Lu P, Yang B, Yang Y, Zhou H, Yang M. Single-cell RNA sequencing analysis of peripheral blood mononuclear cells in PD-1-induced renal toxicity in patients with lung cancer. BMC Nephrol 2024; 25:307. [PMID: 39277735 PMCID: PMC11401319 DOI: 10.1186/s12882-024-03754-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/11/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Although the patient survival rate for many malignancies has been improved with immune checkpoint inhibitors (ICIs), some patients experience various immune-related adverse events (irAEs). IrAEs impact several organ systems, including the kidney. With anti-programmed cell death protein 1 (PD-1) therapy (pembrolizumab), kidney-related adverse events occur relatively rarely compared with other irAEs. However, the occurrence of AKI usually leads to anti-PD-1 therapy interruption or discontinuation. Therefore, there is an urgent need to clarify the mechanisms of renal irAEs (R-irAEs) to facilitate early management. This study aimed to analyse the characteristics of peripheral blood mononuclear cells (PBMCs) in R-irAEs. METHODS PBMCs were collected from three patients who developed R-irAEs after anti-PD-1 therapy and three patients who did not. The PBMCs were subjected to scRNA-seq to identify cell clusters and differentially expressed genes (DEGs). Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analyses were performed to investigate the most active biological processes in immune cells. RESULTS Fifteen cell clusters were identified across the two groups. FOS, RPS26, and JUN were the top three upregulated genes in CD4+ T cells. The DEGs in CD4+ T cells were enriched in Th17 differentiation, Th1 and Th2 cell differentiation, NF-kappa B, Nod-like receptor, TNF, IL-17, apoptosis, and NK cell-mediated cytotoxicity signaling pathways. RPS26, TRBV25-1, and JUN were the top three upregulated genes in CD8+ T cells. The DEGs in CD8+ T cells were enriched in Th17 cell differentiation, antigen processing and presentation, natural killer cell-mediated cytotoxicity, the intestinal immune network for IgA production, the T-cell receptor signalling pathway, Th1 and Th2 cell differentiation, the phagosome, and cell adhesion molecules. CONCLUSIONS In conclusion, R-irAEs are associated with immune cell dysfunction. DEGs and their enriched pathways identified in CD4+ T cells and CD8+ T cells play important roles in the development of renal irAEs related to anti-PD-1 therapy. These findings offer fresh perspectives on the pathogenesis of renal damage caused by anti-PD-1 therapy.
Collapse
Affiliation(s)
- Shusu Liu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Peiyu Lu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Bixia Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Yan Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China.
| |
Collapse
|
3
|
Wang X, Sun Z, Fu J, Fang Z, Zhang W, He JC, Lee K. LRG1 loss effectively restrains glomerular TGF-β signaling to attenuate diabetic kidney disease. Mol Ther 2024; 32:3177-3193. [PMID: 38910328 PMCID: PMC11403230 DOI: 10.1016/j.ymthe.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Transforming growth factor (TGF)-β signaling is a well-established pathogenic mediator of diabetic kidney disease (DKD). However, owing to its pleiotropic actions, its systemic blockade is not therapeutically optimal. The expression of TGF-β signaling regulators can substantially influence TGF-β's effects in a cell- or context-specific manner. Among these, leucine-rich α2-glycoprotein 1 (LRG1) is significantly increased in glomerular endothelial cells (GECs) in DKD. As LRG1 is a secreted molecule that can exert autocrine and paracrine effects, we examined the effects of LRG1 loss in kidney cells in diabetic OVE26 mice by single-cell transcriptomic analysis. Gene expression analysis confirmed a predominant expression of Lrg1 in GECs, which further increased in diabetic kidneys. Loss of Lrg1 led to the reversal of angiogenic and TGF-β-induced gene expression in GECs, which were associated with DKD attenuation. Notably, Lrg1 loss also mitigated the increased TGF-β-mediated gene expression in both podocytes and mesangial cells in diabetic mice, indicating that GEC-derived LRG1 potentiates TGF-β signaling in glomerular cells in an autocrine and paracrine manner. Indeed, a significant reduction in phospho-Smad proteins was observed in the glomerular cells of OVE26 mice with LRG1 loss. These results indicate that specific antagonisms of LRG1 may be an effective approach to curb the hyperactive glomerular TGF-β signaling to attenuate DKD.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeguo Sun
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jia Fu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengying Fang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Weijia Zhang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, NY 10468, USA.
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Sánchez-Ceinos J, Hussain S, Khan AW, Zhang L, Almahmeed W, Pernow J, Cosentino F. Repressive H3K27me3 drives hyperglycemia-induced oxidative and inflammatory transcriptional programs in human endothelium. Cardiovasc Diabetol 2024; 23:122. [PMID: 38580969 PMCID: PMC10998410 DOI: 10.1186/s12933-024-02196-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Histone modifications play a critical role in chromatin remodelling and regulate gene expression in health and disease. Histone methyltransferases EZH1, EZH2, and demethylases UTX, JMJD3, and UTY catalyse trimethylation of lysine 27 on histone H3 (H3K27me3). This study was designed to investigate whether H3K27me3 triggers hyperglycemia-induced oxidative and inflammatory transcriptional programs in the endothelium. METHODS We studied human aortic endothelial cells exposed to high glucose (HAEC) or isolated from individuals with diabetes (D-HAEC). RT-qPCR, immunoblotting, chromatin immunoprecipitation (ChIP-qPCR), and confocal microscopy were performed to investigate the role of H3K27me3. We determined superoxide anion (O2-) production by ESR spectroscopy, NF-κB binding activity, and monocyte adhesion. Silencing/overexpression and pharmacological inhibition of chromatin modifying enzymes were used to modulate H3K27me3 levels. Furthermore, isometric tension studies and immunohistochemistry were performed in aorta from wild-type and db/db mice. RESULTS Incubation of HAEC to high glucose showed that upregulation of EZH2 coupled to reduced demethylase UTX and JMJD3 was responsible for the increased H3K27me3. ChIP-qPCR revealed that repressive H3K27me3 binding to superoxide dismutase and transcription factor JunD promoters is involved in glucose-induced O2- generation. Indeed, loss of JunD transcriptional inhibition favours NOX4 expression. Furthermore, H3K27me3-driven oxidative stress increased NF-κB p65 activity and downstream inflammatory genes. Interestingly, EZH2 inhibitor GSK126 rescued these endothelial derangements by reducing H3K27me3. We also found that H3K27me3 epigenetic signature alters transcriptional programs in D-HAEC and aortas from db/db mice. CONCLUSIONS EZH2-mediated H3K27me3 represents a key epigenetic driver of hyperglycemia-induced endothelial dysfunction. Targeting EZH2 may attenuate oxidative stress and inflammation and, hence, prevent vascular disease in diabetes.
Collapse
Affiliation(s)
- Julia Sánchez-Ceinos
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Shafaat Hussain
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Abdul Waheed Khan
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Liang Zhang
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - John Pernow
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
5
|
Ma S, Xie X, Yuan R, Xin Q, Miao Y, Leng SX, Chen K, Cong W. Vascular Aging and Atherosclerosis: A Perspective on Aging. Aging Dis 2024:AD.2024.0201-1. [PMID: 38502584 DOI: 10.14336/ad.2024.0201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular aging (VA) is recognized as a pivotal factor in the development and progression of atherosclerosis (AS). Although various epidemiological and clinical research has demonstrated an intimate connection between aging and AS, the candidate mechanisms still require thorough examination. This review adopts an aging-centric perspective to deepen the comprehension of the intricate relationship between biological aging, vascular cell senescence, and AS. Various aging-related physiological factors influence the physical system's reactions, including oxygen radicals, inflammation, lipids, angiotensin II, mechanical forces, glucose levels, and insulin resistance. These factors cause endothelial dysfunction, barrier damage, sclerosis, and inflammation for VA and promote AS via distinct or shared pathways. Furthermore, the increase of senescent cells inside the vascular tissues, caused by genetic damage, dysregulation, secretome changes, and epigenetic modifications, might be the primary cause of VA.
Collapse
Affiliation(s)
- Shudong Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuena Xie
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Sean Xiao Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Keji Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weihong Cong
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
6
|
Yin J, Fu X, Luo Y, Leng Y, Ao L, Xie C. A Narrative Review of Diabetic Macroangiopathy: From Molecular Mechanism to Therapeutic Approaches. Diabetes Ther 2024; 15:585-609. [PMID: 38302838 PMCID: PMC10942953 DOI: 10.1007/s13300-024-01532-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Diabetic macroangiopathy, a prevalent and severe complication of diabetes mellitus, significantly contributes to the increased morbidity and mortality rates among affected individuals. This complex disorder involves multifaceted molecular mechanisms that lead to the dysfunction and damage of large blood vessels, including atherosclerosis (AS) and peripheral arterial disease. Understanding the intricate pathways underlying the development and progression of diabetic macroangiopathy is crucial for the development of effective therapeutic interventions. This review aims to shed light on the molecular mechanism implicated in the pathogenesis of diabetic macroangiopathy. We delve into the intricate interplay of chronic inflammation, oxidative stress, endothelial dysfunction, and dysregulated angiogenesis, all of which contribute to the vascular complications observed in this disorder. By exploring the molecular mechanism involved in the disease we provide insight into potential therapeutic targets and strategies. Moreover, we discuss the current therapeutic approaches used for treating diabetic macroangiopathy, including glycemic control, lipid-lowering agents, and vascular interventions.
Collapse
Affiliation(s)
- Jiacheng Yin
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Xiaoxu Fu
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yue Luo
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yuling Leng
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Lianjun Ao
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
7
|
Miao Y, Zhang S, Liang Z, Wang Y, Tian D, Jin S, Guo Q, Xue H, Teng X, Xiao L, Wu Y. Hydrogen sulfide ameliorates endothelial dysfunction in aging arteries by regulating ferroptosis. Nitric Oxide 2023; 140-141:77-90. [PMID: 37875241 DOI: 10.1016/j.niox.2023.10.002] [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: 08/28/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 10/26/2023]
Abstract
Aging causes vascular endothelial dysfunction. We aimed to investigate the causes of vascular endothelial dysfunction during aging using plasma and renal arteries from patients who underwent nephrectomy and animal models. The results showed that the endogenous H2S-producing enzyme cystathione-γ-lyase (CSE) protein expression was downregulated in renal artery tissue, plasma H2S levels were reduced. Moreover, elevated lipid peroxidation and iron accumulation levels led to ferroptosis and endothelial diastolic function in the renal arteries was impaired in the elderly group. H2S enhanced the endogenous CSE expression in the elderly group, promoted endogenous H2S production, decreased lipid peroxide expression, and inhibited ferroptosis, which in turn improved vascular endothelial function in the elderly group. In animal models, we also observed the same results. In addition, we applied NaHS, Ferrostatin-1 (ferroptosis inhibitor) and erastin (ferroptosis inducer) to incubate renal arteries of SD rats. The results showed that NaHS enhanced ferroptosis related proteins expression, inhibited ferroptosis and improved vascular endothelial function. We demonstrated that endothelial dysfunction associated with aging is closely related to reduced endogenous H2S levels and ferroptosis in vascular endothelial cells. Notably, H2S reduced lipid peroxidation levels in vascular endothelial cells, inhibited ferroptosis in vascular endothelial cells, and improved endothelial dysfunction.
Collapse
Affiliation(s)
- Yuxin Miao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Shuangshuang Zhang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Zihui Liang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Yang Wang
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Danyang Tian
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Hongmei Xue
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Xu Teng
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China.
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China; Experimental Center for Teaching, Hebei Medical University, Shijiazhuang, 050017, China; Key Laboratory of Vascular Medicine of Hebei Province, Shijiazhuang, 050017, China.
| |
Collapse
|
8
|
Casper E. The crosstalk between Nrf2 and NF-κB pathways in coronary artery disease: Can it be regulated by SIRT6? Life Sci 2023; 330:122007. [PMID: 37544377 DOI: 10.1016/j.lfs.2023.122007] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/26/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Oxidative stress and inflammation are major mechanisms responsible for the progression of CAD. Nuclear transcription factor erythroid-2 related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox status. Nrf2 upregulation increases the expression of antioxidant genes, decreases the expression of Nuclear factor-kappa B (NF-kB), and increases free radical metabolism. Activated NF-kB increases the production of inflammatory cytokines causing endothelial dysfunction. The two pathways of Nrf2 and NF-kB can regulate the expression of each other. Foremost, the Nrf2 pathway can decrease the level of active NF-κB by increasing the level of antioxidants and cytoprotective enzymes. Furthermore, the Nrf2 pathway prevents IκB-α degradation, an inhibitor of NF-kB, and thus inhibits NF-κB mediated transcription. Also, NF-kB transcription inhibits Nrf2 activation by reducing the antioxidant response element (ARE) transcription. Sirtuin 6 (SIRT6) is a member of the Sirtuins family that was found to protect against cardiovascular diseases. SIRT6 can suppress the production of Reactive oxygen species (ROS) through deacetylation of NRF2 which results in NRF2 activation. Furthermore, SIRT6 can inhibit the inflammatory process through the downregulation of NF-kB transcription. Therefore, targeting sirtuins could be a therapeutic strategy to treat CAD. This review describes the potential role of SIRT6 in regulating the crosstalk between NRF2 and NF-kB signaling pathways in CAD.
Collapse
Affiliation(s)
- Eman Casper
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
9
|
Ujiie N, Norden PR, Fang R, Beckmann L, Cai Z, Kweon J, Liu T, Tan C, Kuhn MS, Stamer WD, Aoto K, Quaggin SE, Zhang HF, Kume T. Differential roles of FOXC2 in the trabecular meshwork and Schlemm's canal in glaucomatous pathology. Life Sci Alliance 2023; 6:e202201721. [PMID: 37414529 PMCID: PMC10326420 DOI: 10.26508/lsa.202201721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Impaired development and maintenance of Schlemm's canal (SC) are associated with perturbed aqueous humor outflow and intraocular pressure. The angiopoietin (ANGPT)/TIE2 signaling pathway regulates SC development and maintenance, whereas the molecular mechanisms of crosstalk between SC and the neural crest (NC)-derived neighboring tissue, the trabecular meshwork (TM), are poorly understood. Here, we show NC-specific forkhead box (Fox)c2 deletion in mice results in impaired SC morphogenesis, loss of SC identity, and elevated intraocular pressure. Visible-light optical coherence tomography analysis further demonstrated functional impairment of the SC in response to changes in intraocular pressure in NC-Foxc2 -/- mice, suggesting altered TM biomechanics. Single-cell RNA-sequencing analysis identified that this phenotype is predominately characterized by transcriptional changes associated with extracellular matrix organization and stiffness in TM cell clusters, including increased matrix metalloproteinase expression, which can cleave the TIE2 ectodomain to produce soluble TIE2. Moreover, endothelial-specific Foxc2 deletion impaired SC morphogenesis because of reduced TIE2 expression, which was rescued by deleting the TIE2 phosphatase VE-PTP. Thus, Foxc2 is critical in maintaining SC identity and morphogenesis via TM-SC crosstalk.
Collapse
Affiliation(s)
- Naoto Ujiie
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Pieter R Norden
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Raymond Fang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Lisa Beckmann
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Zhen Cai
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Junghun Kweon
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Ting Liu
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Can Tan
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Megan S Kuhn
- Duke Eye Center, Duke University, Durham, NC, USA
| | | | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
10
|
Sun N, Akay LA, Murdock MH, Park Y, Galiana-Melendez F, Bubnys A, Galani K, Mathys H, Jiang X, Ng AP, Bennett DA, Tsai LH, Kellis M. Single-nucleus multiregion transcriptomic analysis of brain vasculature in Alzheimer's disease. Nat Neurosci 2023; 26:970-982. [PMID: 37264161 PMCID: PMC10464935 DOI: 10.1038/s41593-023-01334-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/17/2023] [Indexed: 06/03/2023]
Abstract
Cerebrovascular dysregulation is a hallmark of Alzheimer's disease (AD), but the changes that occur in specific cell types have not been fully characterized. Here, we profile single-nucleus transcriptomes in the human cerebrovasculature in six brain regions from 220 individuals with AD and 208 age-matched controls. We annotate 22,514 cerebrovascular cells, including 11 subtypes of endothelial, pericyte, smooth muscle, perivascular fibroblast and ependymal cells. We identify 2,676 differentially expressed genes in AD, including downregulation of PDGFRB in pericytes, and of ABCB1 and ATP10A in endothelial cells, and validate the downregulation of SLC6A1 and upregulation of APOD, INSR and COL4A1 in postmortem AD brain tissues. We detect vasculature, glial and neuronal coexpressed gene modules, suggesting coordinated neurovascular unit dysregulation in AD. Integration with AD genetics reveals 125 AD differentially expressed genes directly linked to AD-associated genetic variants. Lastly, we show that APOE4 genotype-associated differences are significantly enriched among AD-associated genes in capillary and venule endothelial cells, as well as subsets of pericytes and fibroblasts.
Collapse
Affiliation(s)
- Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Leyla Anne Akay
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mitchell H Murdock
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yongjin Park
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology and Laboratory Medicine, Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Fabiola Galiana-Melendez
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adele Bubnys
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kyriaki Galani
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xueqiao Jiang
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ayesha P Ng
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Li-Huei Tsai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Mengozzi A, Costantino S, Mongelli A, Mohammed SA, Gorica E, Delfine V, Masi S, Virdis A, Ruschitzka F, Paneni F. Epigenetic Signatures in Arterial Hypertension: Focus on the Microvasculature. Int J Mol Sci 2023; 24:ijms24054854. [PMID: 36902291 PMCID: PMC10003673 DOI: 10.3390/ijms24054854] [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: 01/31/2023] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Systemic arterial hypertension (AH) is a multifaceted disease characterized by accelerated vascular aging and high cardiometabolic morbidity and mortality. Despite extensive work in the field, the pathogenesis of AH is still incompletely understood, and its treatment remains challenging. Recent evidence has shown a deep involvement of epigenetic signals in the regulation of transcriptional programs underpinning maladaptive vascular remodeling, sympathetic activation and cardiometabolic alterations, all factors predisposing to AH. After occurring, these epigenetic changes have a long-lasting effect on gene dysregulation and do not seem to be reversible upon intensive treatment or the control of cardiovascular risk factors. Among the factors involved in arterial hypertension, microvascular dysfunction plays a central role. This review will focus on the emerging role of epigenetic changes in hypertensive-related microvascular disease, including the different cell types and tissues (endothelial cells, vascular smooth muscle cells and perivascular adipose tissue) as well as the involvement of mechanical/hemodynamic factors, namely, shear stress.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Shafeeq A. Mohammed
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, 8952 Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
- Correspondence: or francesco.paneni@uzh; Tel.: +41-44-6355096
| |
Collapse
|
12
|
Lazzeroni D, Villatore A, Souryal G, Pili G, Peretto G. The Aging Heart: A Molecular and Clinical Challenge. Int J Mol Sci 2022; 23:16033. [PMID: 36555671 PMCID: PMC9783309 DOI: 10.3390/ijms232416033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is associated with an increasing burden of morbidity, especially for cardiovascular diseases (CVDs). General cardiovascular risk factors, ischemic heart diseases, heart failure, arrhythmias, and cardiomyopathies present a significant prevalence in older people, and are characterized by peculiar clinical manifestations that have distinct features compared with the same conditions in a younger population. Remarkably, the aging heart phenotype in both healthy individuals and patients with CVD reflects modifications at the cellular level. An improvement in the knowledge of the physiological and pathological molecular mechanisms underlying cardiac aging could improve clinical management of older patients and offer new therapeutic targets.
Collapse
Affiliation(s)
| | - Andrea Villatore
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Department of Arrhythmology and Cardiac Electrophysiology, Ospedale San Raffaele, 20132 Milan, Italy
| | - Gaia Souryal
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Gianluca Pili
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Giovanni Peretto
- School of Medicine, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Department of Arrhythmology and Cardiac Electrophysiology, Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
13
|
Mengozzi A, Costantino S, Paneni F, Duranti E, Nannipieri M, Mancini R, Lai M, La Rocca V, Puxeddu I, Antonioli L, Fornai M, Ghionzoli M, Georgiopoulos G, Ippolito C, Bernardini N, Ruschitzka F, Pugliese NR, Taddei S, Virdis* A, Masi S. Targeting SIRT1 Rescues Age- and Obesity-Induced Microvascular Dysfunction in Ex Vivo Human Vessels. Circ Res 2022; 131:476-491. [PMID: 35968712 PMCID: PMC9426744 DOI: 10.1161/circresaha.122.320888] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Experimental evidence suggests a key role of SIRT1 (silent information regulator 1) in age- and metabolic-related vascular dysfunction. Whether these effects hold true in the human microvasculature is unknown. We aimed to investigate the SIRT1 role in very early stages of age- and obesity-related microvascular dysfunction in humans. METHODS Ninety-five subjects undergoing elective laparoscopic surgery were recruited and stratified based on their body mass index status (above or below 30 kg/m2) and age (above or below 40 years) in 4 groups: Young Nonobese, Young Obese, Old Nonobese, and Old Obese. We measured small resistance arteries' endothelial function by pressurized micromyography before and after incubation with a SIRT1 agonist (SRT1720) and a mitochondria reactive oxygen species (mtROS) scavenger (MitoTEMPO). We assessed vascular levels of mtROS and nitric oxide availability by confocal microscopy and vascular gene expression of SIRT1 and mitochondrial proteins by qPCR. Chromatin immunoprecipitation assay was employed to investigate SIRT1-dependent epigenetic regulation of mitochondrial proteins. RESULTS Compared with Young Nonobese, obese and older patients showed lower vascular expression of SIRT1 and antioxidant proteins (FOXO3 [forkhead box protein O3] and SOD2) and higher expression of pro-oxidant and aging mitochondria proteins p66Shc and Arginase II. Old Obese, Young Obese and Old Nonobese groups endothelial dysfunction was rescued by SRT1720. The restoration was comparable to the one obtained with mitoTEMPO. These effects were explained by SIRT1-dependent chromatin changes leading to reduced p66Shc expression and upregulation of proteins involved in mitochondria respiratory chain. CONCLUSIONS SIRT1 is a novel central modulator of the earliest microvascular damage induced by age and obesity. Through a complex epigenetic control mainly involving p66Shc and Arginase II, it influences mtROS levels, NO availability, and the expression of proteins of the mitochondria respiratory chain. Therapeutic modulation of SIRT1 restores obesity- and age-related endothelial dysfunction. Early targeting of SIRT1 might represent a crucial strategy to prevent age- and obesity-related microvascular dysfunction.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Switzerland (S.C., F.P.)
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Switzerland (S.C., F.P.).,Department of Cardiology, University Heart Center (F.P., F.R.), University Hospital Zurich, Switzerland.,Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Emiliano Duranti
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Rudj Mancini
- Unit of Bariatric Surgery, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy (R.M.)
| | - Michele Lai
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery (M.L., V.L.R.), University of Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery (M.L., V.L.R.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Marco Ghionzoli
- Paediatric Surgery Unit, Meyer Children’s Hospital, Florence, Italy (M.G.)
| | - Georgios Georgiopoulos
- School of Biomedical Engineering and Imaging Sciences, King’s College London, United Kingdom (G.G.).,Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Greece (G.G.)
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center (F.P., F.R.), University Hospital Zurich, Switzerland
| | - Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Agostino Virdis*
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
| |
Collapse
|
14
|
The Level of HbA1c Evaluates the Extent of Coronary Atherosclerosis Lesions and the Prognosis in Diabetes with Acute Coronary Syndrome. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7796809. [PMID: 35912151 PMCID: PMC9337927 DOI: 10.1155/2022/7796809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/04/2022] [Accepted: 03/12/2022] [Indexed: 11/17/2022]
Abstract
Background The level of HbA1c can reflect the average level of blood glucose over 3 months, which is the gold standard indicator for monitoring blood glucose. The relationship between the level of HbA1c and the extent of coronary atherosclerosis lesions or the prognosis in diabetes with acute coronary syndrome (ACS) remains poorly understood. Aims To explore whether the level of HbA1c can evaluate the extent of coronary atherosclerosis lesions or the prognosis in diabetes with acute coronary syndrome (ACS) using the SYNTAX score, the Global Registry of Acute Coronary Events (GRACE) score, left ventricular function (LVEF), left ventricular end-diastolic volume (LVEDV), and major adverse cardiac events (MACEs) in the hospital and 12 months after discharge. Methods This study was a prospective, randomized, open-label, and parallel group study. Patients with diabetes with ACS were recruited into this study indiscriminately, and all the participants were divided into two groups according to the level of HbA1c: HbA1c level ≤ 7%group and HbA1c level > 7%group. The followings were used as the evaluation indicators: SYNTAX score, GRACE score, LVEF, LVEDV, and MACEs in hospital and 12 months after discharge. Results A total of 233 patients with diabetes and ACS were enrolled and assigned to two groups according to their level of HbA1c: the HbA1c ≤ 7%group (n = 92) and the HbA1c > 7%group (n = 141). The results showed that the proportion of STEMI was higher in the HbA1c ≤7% group (p < 0.05), while the proportion of NSTEMI has not significantly higher in the HbA1c >7% group (p > 0.05). Regression analysis indicated that HbA1c level was significantly positively correlated with GRACE score (r = 0.156, F = 5.784, p = 0.017, n = 233) and SYNTAX score (r = 0.237, F = 13.788, p < 0.001, n = 233), and there were no statistically significant differences in LVEDV and LVEF between the two groups (p > 0.05). The total MACEs rate showed no significant difference between the two groups during hospitalization (p > 0.05) but showed significant differences at 12 months after discharge (p < 0.05). Conclusions This study shows that HbA1c level was positively correlated with the extent of coronary atherosclerosis lesions and the prognosis in diabetes with ACS. The higher the HbA1c level is, the more severe the coronary atherosclerotic lesion and the worse the prognosis in diabetes with ACS are.
Collapse
|
15
|
Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J 2021; 41:2974-2982. [PMID: 32006431 DOI: 10.1093/eurheartj/ehz961] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/10/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
The ongoing worldwide increase in life expectancy portends a rising prevalence of age-related cardiovascular (CV) diseases in the coming decades that demands a deeper understanding of their molecular mechanisms. Inflammation has recently emerged as an important contributor for CV disease development. Indeed, a state of chronic sterile low-grade inflammation characterizes older organisms (also known as inflamm-ageing) and participates pivotally in the development of frailty, disability, and most chronic degenerative diseases including age-related CV and cerebrovascular afflictions. Due to chronic activation of inflammasomes and to reduced endogenous anti-inflammatory mechanisms, inflamm-ageing contributes to the activation of leucocytes, endothelial, and vascular smooth muscle cells, thus accelerating vascular ageing and atherosclerosis. Furthermore, inflamm-ageing promotes the development of catastrophic athero-thrombotic complications by enhancing platelet reactivity and predisposing to plaque rupture and erosion. Thus, inflamm-ageing and its contributors or molecular mediators might furnish targets for novel therapeutic strategies that could promote healthy ageing and conserve resources for health care systems worldwide. Here, we discuss recent findings in the pathophysiology of inflamm-ageing, the impact of these processes on the development of age-related CV diseases, results from clinical trials targeting its components and the potential implementation of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, L.go Rosanna Benzi 10, 16132 Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, v.le Benedetto XV 10, 16132 Genoa, Italy
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montreal, Rue Bélanger 5000, Montreal, QC H1T 1C8, Canada
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Francis Street 75, Boston, MA 02115, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren CH-8952, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
16
|
Ma J, Xing B, Cao Y, He X, Bennett KE, Tong C, An C, Hojnacki T, Feng Z, Deng S, Ling S, Xie G, Wu Y, Ren Y, Yu M, Katona BW, Li H, Naji A, Hua X. Menin-regulated Pbk controls high fat diet-induced compensatory beta cell proliferation. EMBO Mol Med 2021; 13:e13524. [PMID: 33821572 PMCID: PMC8103087 DOI: 10.15252/emmm.202013524] [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: 09/28/2020] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic beta cells undergo compensatory proliferation in the early phase of type 2 diabetes. While pathways such as FoxM1 are involved in regulating compensatory beta cell proliferation, given the lack of therapeutics effectively targeting beta cell proliferation, other targetable pathways need to be identified. Herein, we show that Pbk, a serine/threonine protein kinase, is essential for high fat diet (HFD)‐induced beta cell proliferation in vivo using a Pbk kinase deficiency knock‐in mouse model. Mechanistically, JunD recruits menin and HDAC3 complex to the Pbk promoter to reduce histone H3 acetylation, leading to epigenetic repression of Pbk expression. Moreover, menin inhibitor (MI) disrupts the menin–JunD interaction and augments Pbk transcription. Importantly, MI administration increases beta cell proliferation, ameliorating hyperglycemia, and impaired glucose tolerance (IGT) in HFD‐induced diabetic mice. Notably, Pbk is required for the MI‐induced beta cell proliferation and improvement of IGT. Together, these results demonstrate the repressive role of the menin/JunD/Pbk axis in regulating HFD‐induced compensatory beta cell proliferation and pharmacologically regulating this axis may serve as a novel strategy for type 2 diabetes therapy.
Collapse
Affiliation(s)
- Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bowen Xing
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan Cao
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kate E Bennett
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Chao Tong
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiying An
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Taylor Hojnacki
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sunbin Deng
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunbin Ling
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gengchen Xie
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuan Wu
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ming Yu
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hongzhe Li
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ali Naji
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
17
|
OCT angiography metrics predict intradialytic hypotension episodes in chronic hemodialysis patients: a pilot, prospective study. Sci Rep 2021; 11:7202. [PMID: 33785805 PMCID: PMC8009948 DOI: 10.1038/s41598-021-86609-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/12/2021] [Indexed: 12/04/2022] Open
Abstract
In chronic hemodialysis (HD) patients, intradialytic hypotension (IDH) is a complication that increases mortality risk. We run a pilot study to analyzing possible relationships between optical coherence tomography angiography (OCT-A) metrics and IDH with the aim of evaluating if OCT-A could represent a useful tool to stratify the hypotensive risk in dialysis patients. A total of 35 eyes (35 patients) were analyzed. OCT-A was performed before and after a single dialysis session. We performed OCT-A 3 × 3 mm and 6 × 6 mm scanning area focused on the fovea centralis. Patients were then followed up to 30 days (10 HD sessions) and a total of 73 IDHs were recorded, with 12 patients (60%) experiencing at least one IDH. Different OCT-A parameters were reduced after dialysis: central choroid thickness (CCT), 6 × 6 mm foveal whole vessel density (VD) of superficial capillary plexus (SPC) and 6 × 6 mm foveal VD of deep capillary plexus (DCP). At logistic regression analysis, IDH was positively associated with baseline foveal VD of SCP and DCP, while an inverse association was found with the choroid. In Kaplan–Meier analyses of patients categorized according to the ROC-derived optimal thresholds, CCT, the 3 × 3 foveal VD of SCP, the 3 × 3 mm and 6 × 6 mm foveal VD of DCP and the 6 × 6 mm foveal VD of SCP were strongly associated with a higher risk of IDH over the 30-days follow-up. In HD patients, a single OCT-A measurement may represent a non-invasive, rapid tool to evaluate the compliance of vascular bed to HD stress and to stratify the risk of IDH in the short term.
Collapse
|
18
|
Wang W, Liu F, Xu C, Liu Z, Ma J, Gu L, Jiang Z, Hou J. Lactobacillus plantarum 69-2 Combined with Galacto-Oligosaccharides Alleviates d-Galactose-Induced Aging by Regulating the AMPK/SIRT1 Signaling Pathway and Gut Microbiota in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2745-2757. [PMID: 33565862 DOI: 10.1021/acs.jafc.0c06730] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Probiotics and prebiotics for preventing and alleviating the degenerative changes associated with aging have received extensive attention. In the present work, Lactobacillus plantarum (L. plantarum) 69-2 with the highest antioxidant capacity combined with galacto-oligosaccharide (GOS) was used in aging model mice to evaluate the effect on aging and the regulation of gut microbiota. The combination of L. plantarum 69-2 and GOS supplementation could significantly (P < 0.05) improve liver function, antioxidant capacity, and inflammation accompanied by regulating the gut microbiota, increasing the short chain fatty acid (SCFA) levels, and activating the hepatic AMPK/SIRT1 regulatory pathway. The results showed that L. plantarum 69-2 and GOS could activate the hepatic AMPK/SIRT1 signaling pathway by regulating the gut microbiota and metabolites through the liver-gut axis to restore hepatic antioxidant activity to alleviate aging. The study provided a new insight for targeting the gut microbiota to relieve aging through the gut-liver axis.
Collapse
Affiliation(s)
- Wan Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Cong Xu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhijing Liu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiage Ma
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Liya Gu
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhanmei Jiang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Juncai Hou
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
19
|
Zhu SY, Liu LL, Huang YQ, Li XW, Talukder M, Dai XY, Li YH, Li JL. In silico analysis of selenoprotein N (Gallus gallus): absence of EF-hand motif and the role of CUGS-helix domain in antioxidant protection. Metallomics 2021; 13:6132312. [PMID: 33693771 DOI: 10.1093/mtomcs/mfab004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Selenoprotein N (SEPN1) is critical to the normal muscular physiology. Mutation of SEPN1 can raise congenital muscular disorder in human. It is also central to maturation and structure of skeletal muscle in chicken. However, human SEPN1 contained an EF-hand motif, which was not found in chicken. And the biochemical and molecular characterization of chicken SEPN1 remains unclear. Hence, protein domains, transcription factors, and interactions of Ca2+ in SEPN1 were analyzed in silico to provide the divergence and homology between chicken and human in this work. The results showed that vertebrates' SEPN1 evolved from a common ancestor. Human and chicken's SEPN1 shared a conserved CUGS-helix domain with function in antioxidant protection. SEPN1 might be a downstream target of JNK pathway, and it could respond to multiple stresses. Human's SEPN1 might not combine with Ca2+ with a single EF-hand motif in calcium homeostasis, and chicken SEPN1 did not have the EF-hand motif in the prediction, indicating the EF-hand motif malfunctioned in chicken SEPN1.
Collapse
Affiliation(s)
- Shi-Yong Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Li-Li Liu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, P. R. China
| | - Yue-Qiang Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xiao-Wei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Milton Talukder
- Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Xue-Yan Dai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.,Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P. R. China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
20
|
Abstract
Emerging evidence suggests the growing importance of "nongenetic factors" in the pathogenesis of atherosclerotic vascular disease. Indeed, the inherited genome determines only part of the risk profile as genomic approaches do not take into account additional layers of biological regulation by "epi"-genetic changes. Epigenetic modifications are defined as plastic chemical changes of DNA/histone complexes which critically affect gene activity without altering the DNA sequence. These modifications include DNA methylation, histone posttranslational modifications, and non-coding RNAs and have the ability to modulate gene expression at both transcriptional and posttranscriptional level. Notably, epigenetic signals are mainly induced by environmental factors (i.e., pollution, smoking, noise) and, once acquired, may be transmitted to the offspring. The inheritance of adverse epigenetic changes may lead to premature deregulation of pathways involved in vascular damage and endothelial dysfunction. Here, we describe the emerging role of epigenetic modifications as fine-tuners of gene transcription in atherosclerosis. Specifically, the following aspects are described in detail: (1) discovery and impact of the epigenome in cardiovascular disease, (2) the epigenetic landscape in atherosclerosis; (3) inheritance of epigenetic signals and premature vascular disease; (4) epigenetic control of lipid metabolism, vascular oxidative stress, inflammation, autophagy, and apoptosis; (5) epigenetic biomarkers in patients with atherosclerosis; (6) novel therapeutic strategies to modulate epigenetic marks. Understanding the individual epigenetic profile may pave the way for new approaches to determine cardiovascular risk and to develop personalized therapies to treat atherosclerosis and its complications.
Collapse
|
21
|
Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life (Basel) 2021; 11:60. [PMID: 33467601 PMCID: PMC7829951 DOI: 10.3390/life11010060] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging can be seen as process characterized by accumulation of oxidative stress induced damage. Oxidative stress derives from different endogenous and exogenous processes, all of which ultimately lead to progressive loss in tissue and organ structure and functions. The oxidative stress theory of aging expresses itself in age-related diseases. Aging is in fact a primary risk factor for many diseases and in particular for cardiovascular diseases and its derived morbidity and mortality. Here we highlight the role of oxidative stress in age-related cardiovascular aging and diseases. We take into consideration the molecular mechanisms, the structural and functional alterations, and the diseases accompanied to the cardiovascular aging process.
Collapse
Affiliation(s)
- Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paolo Vitillo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
22
|
Single-cell RNA sequencing in vision research: Insights into human retinal health and disease. Prog Retin Eye Res 2020; 83:100934. [PMID: 33383180 DOI: 10.1016/j.preteyeres.2020.100934] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/03/2023]
Abstract
Gene expression provides valuable insight into cell function. As such, vision researchers have frequently employed gene expression studies to better understand retinal physiology and disease. With the advent of single-cell RNA sequencing, expression experiments provide an unparalleled resolution of information. Instead of studying aggregated gene expression across all cells in a heterogenous tissue, single-cell technology maps RNA to an individual cell, which facilitates grouping of retinal and choroidal cell types for further study. Single-cell RNA sequencing has been quickly adopted by both basic and translational vision researchers, and single-cell level gene expression has been studied in the visual systems of animal models, retinal organoids, and primary human retina, RPE, and choroid. These experiments have generated detailed atlases of gene expression and identified new retinal cell types. Likewise, single-cell RNA sequencing investigations have characterized how gene expression changes in the setting of many retinal diseases, including how choroidal endothelial cells are altered in age-related macular degeneration. In addition, this technology has allowed vision researchers to discover drivers of retinal development and model rare retinal diseases with induced pluripotent stem cells. In this review, we will overview the growing number of single-cell RNA sequencing studies in the field of vision research. We will summarize experimental considerations for designing single-cell RNA sequencing experiments and highlight important advancements in retinal, RPE, choroidal, and retinal organoid biology driven by this technology. Finally, we generalize these findings to genes involved in retinal degeneration and outline the future of single-cell expression experiments in studying retinal disease.
Collapse
|
23
|
Regnault V, Challande P, Pinet F, Li Z, Lacolley P. Cell senescence: basic mechanisms and the need for computational networks in vascular ageing. Cardiovasc Res 2020; 117:1841-1858. [PMID: 33206947 DOI: 10.1093/cvr/cvaa318] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/26/2020] [Accepted: 10/28/2020] [Indexed: 01/10/2023] Open
Abstract
This review seeks to provide an update of the mechanisms of vascular cell senescence, from newly identified molecules to arterial ageing phenotypes, and finally to present a computational approach to connect these selected proteins in biological networks. We will discuss current key signalling and gene expression pathways by which these focus proteins and networks drive normal and accelerated vascular ageing. We also review the possibility that senolytic drugs, designed to restore normal cell differentiation and function, could effectively treat multiple age-related vascular diseases. Finally, we discuss how cell senescence is both a cause and a consequence of vascular ageing because of the possible feedback controls between identified networks.
Collapse
Affiliation(s)
- Véronique Regnault
- Université de Lorraine, INSERM, DCAC, 9 avenue de la forêt de Haye, CS 50184, 54000 Nancy, France
| | - Pascal Challande
- Sorbonne Université, CNRS, Institut Jean Le Rond d'Alembert, 4 place Jussieu, 75005 Paris, France
| | - Florence Pinet
- Univ. Lille, CHU Lille, Inserm, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France
| | - Zhenlin Li
- Sorbonne Université, CNRS, INSERM, IBPS, Biological Adaptation and Aging, Paris, France
| | - Patrick Lacolley
- Université de Lorraine, INSERM, DCAC, 9 avenue de la forêt de Haye, CS 50184, 54000 Nancy, France
| |
Collapse
|
24
|
Hussain S, Khan AW, Akhmedov A, Suades R, Costantino S, Paneni F, Caidahl K, Mohammed SA, Hage C, Gkolfos C, Björck H, Pernow J, Lund LH, Lüscher TF, Cosentino F. Hyperglycemia Induces Myocardial Dysfunction via Epigenetic Regulation of JunD. Circ Res 2020; 127:1261-1273. [PMID: 32815777 DOI: 10.1161/circresaha.120.317132] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
RATIONALE Hyperglycemia -induced reactive oxygen species are key mediators of cardiac dysfunction. JunD (Jund proto-oncogene subunit), a member of the AP-1 (activator protein-1) family of transcription factors, is emerging as a major gatekeeper against oxidative stress. However, its contribution to redox state and inflammation in the diabetic heart remains to be elucidated. OBJECTIVE The present study investigates the role of JunD in hyperglycemia-induced and reactive oxygen species-driven myocardial dysfunction. METHODS AND RESULTS JunD mRNA and protein expression were reduced in the myocardium of mice with streptozotocin-induced diabetes mellitus as compared to controls. JunD downregulation was associated with oxidative stress and left ventricular dysfunction assessed by electron spin resonance spectroscopy as well as conventional and 2-dimensional speckle-tracking echocardiography. Furthermore, myocardial expression of free radical scavenger superoxide dismutase 1 and aldehyde dehydrogenase 2 was reduced, whereas the NOX2 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 2) and NOX4 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 4) were upregulated. The redox changes were associated with increased NF-κB (nuclear factor kappa B) binding activity and expression of inflammatory mediators. Interestingly, mice with cardiac-specific overexpression of JunD via the α MHC (α- myosin heavy chain) promoter (α MHC JunDtg) were protected against hyperglycemia-induced cardiac dysfunction. We also showed that JunD was epigenetically regulated by promoter hypermethylation, post-translational modification of histone marks, and translational repression by miRNA (microRNA)-673/menin. Reduced JunD mRNA and protein expression were confirmed in left ventricular specimens obtained from patients with type 2 diabetes mellitus as compared to nondiabetic subjects. CONCLUSIONS Here, we show that a complex epigenetic machinery involving DNA methylation, histone modifications, and microRNAs mediates hyperglycemia-induced JunD downregulation and myocardial dysfunction in experimental and human diabetes mellitus. Our results pave the way for tissue-specific therapeutic modulation of JunD to prevent diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shafaat Hussain
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Abdul Waheed Khan
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Switzerland (A.A., S.C., F.P., S.A.M., T.F.L.)
| | - Rosa Suades
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zurich, Switzerland (A.A., S.C., F.P., S.A.M., T.F.L.)
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zurich, Switzerland (A.A., S.C., F.P., S.A.M., T.F.L.).,University Heart Center and Department of Research and Education, University Hospital Zürich, Switzerland (F.P.)
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery (K.C.), Karolinska Institutet, Stockholm, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden (K.C.)
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zurich, Switzerland (A.A., S.C., F.P., S.A.M., T.F.L.)
| | - Camilla Hage
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Christos Gkolfos
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Hanna Björck
- Center for Molecular Medicine, Department of Medicine (H.B.), Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Lars H Lund
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland (A.A., S.C., F.P., S.A.M., T.F.L.)
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (S.H., A.W.K., R.S., C.H., C.G., J.P., L.H.L., F.C.)
| |
Collapse
|
25
|
Poznyak AV, Grechko AV, Orekhova VA, Khotina V, Ivanova EA, Orekhov AN. NADPH Oxidases and Their Role in Atherosclerosis. Biomedicines 2020; 8:biomedicines8070206. [PMID: 32664404 PMCID: PMC7399834 DOI: 10.3390/biomedicines8070206] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
The current view on atherosclerosis positions it as a multifactorial disorder that results from the interplay between lipid metabolism disturbances and inflammatory processes. Oxidative stress is proven to be one of the initiating factors in atherosclerosis development, being implicated both in the inflammatory response and in atherogenic modifications of lipoproteins that facilitate lipid accumulation in the arterial wall. The hallmark of oxidative stress is the elevated level of reactive oxygen species (ROS). Correspondingly, the activity of major ROS-generating enzymes, including nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, and cyclooxygenases, is an important element in atherosclerosis development. In particular, the role of NADPH oxidases in atherosclerosis development has become a subject of intensive research. Aberrant activity of NADPH oxidases was shown to be associated with cardiovascular disease in humans. With regard to atherosclerosis, several important pathological components of the disease development, including endothelial dysfunction, inflammation, and vascular remodeling, involve aberrations in NADPH oxidases functioning. In humans, NADPH oxidases are represented by four isoforms expressed in vascular tissues, where they serve as the main source of ROS during atherogenesis. Moreover, recent studies have demonstrated their impact on vascular remodeling processes. Interestingly, one of the NADPH oxidase isoforms, NOX4, was shown to have an atheroprotective effect. Despite the growing evidence of the crucial involvement of NADPH oxidases in atherosclerosis pathogenesis, the available data still remains controversial. In this narrative review, we summarize the current knowledge of the role of NADPH oxidases in atherosclerosis and outline the future directions of research.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia; (A.V.P.); (E.A.I.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Varvara A. Orekhova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia;
| | - Victoria Khotina
- Laboratory of Infectious Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
| | - Ekaterina A. Ivanova
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia; (A.V.P.); (E.A.I.)
| | - Alexander N. Orekhov
- Laboratory of Infectious Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8, Baltiyskaya St., 125315 Moscow, Russia
- Correspondence: ; Tel./Fax: +7-(495)-4159594
| |
Collapse
|
26
|
Voigt AP, Whitmore SS, Mulfaul K, Chirco KR, Giacalone JC, Flamme-Wiese MJ, Stockman A, Stone EM, Tucker BA, Scheetz TE, Mullins RF. Bulk and single-cell gene expression analyses reveal aging human choriocapillaris has pro-inflammatory phenotype. Microvasc Res 2020; 131:104031. [PMID: 32531351 DOI: 10.1016/j.mvr.2020.104031] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
The human choroidal vasculature is subject to age-related structural and gene expression changes implicated in age-related macular degeneration (AMD). In this study, we performed both bulk and single-cell RNA sequencing on infant (n = 4 for bulk experiments, n = 2 for single-cell experiments) and adult (n = 13 for bulk experiments, n = 6 for single-cell experiments) human donors to characterize how choroidal gene expression changes with age. Differential expression analysis revealed that aged choroidal samples were enriched in genes encoding pro-inflammatory transcription factors and leukocyte transendothelial cell migration adhesion proteins. Such genes were observed to be differentially expressed specifically within choroidal endothelial cells at the single-cell level. Immunohistochemistry experiments support transcriptional findings that CD34 is elevated in infant choriocapillaris endothelial cells while ICAM-1 is enriched in adults. These results suggest several potential drivers of the pro-inflammatory vascular phenotype observed with advancing age.
Collapse
Affiliation(s)
- Andrew P Voigt
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - S Scott Whitmore
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Kelly Mulfaul
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Kathleen R Chirco
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Joseph C Giacalone
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Miles J Flamme-Wiese
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Adam Stockman
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Iowa Lions Eye Bank, Coralville, IA 52241, United States of America
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Budd A Tucker
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Todd E Scheetz
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America
| | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, the University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States of America; Institute for Vision Research, the University of Iowa, Iowa City, IA 52242, United States of America.
| |
Collapse
|
27
|
Liberale L, Camici GG. The Role of Vascular Aging in Atherosclerotic Plaque Development and Vulnerability. Curr Pharm Des 2020; 25:3098-3111. [PMID: 31470777 DOI: 10.2174/1381612825666190830175424] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The ongoing demographical shift is leading to an unprecedented aging of the population. As a consequence, the prevalence of age-related diseases, such as atherosclerosis and its thrombotic complications is set to increase in the near future. Endothelial dysfunction and vascular stiffening characterize arterial aging and set the stage for the development of cardiovascular diseases. Atherosclerotic plaques evolve over time, the extent to which these changes might affect their stability and predispose to sudden complications remains to be determined. Recent advances in imaging technology will allow for longitudinal prospective studies following the progression of plaque burden aimed at better characterizing changes over time associated with plaque stability or rupture. Oxidative stress and inflammation, firmly established driving forces of age-related CV dysfunction, also play an important role in atherosclerotic plaque destabilization and rupture. Several genes involved in lifespan determination are known regulator of redox cellular balance and pre-clinical evidence underlines their pathophysiological roles in age-related cardiovascular dysfunction and atherosclerosis. OBJECTIVE The aim of this narrative review is to examine the impact of aging on arterial function and atherosclerotic plaque development. Furthermore, we report how molecular mechanisms of vascular aging might regulate age-related plaque modifications and how this may help to identify novel therapeutic targets to attenuate the increased risk of CV disease in elderly people.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland.,University Heart Center, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
28
|
Bersini S, Arrojo E Drigo R, Huang L, Shokhirev MN, Hetzer MW. Transcriptional and Functional Changes of the Human Microvasculature during Physiological Aging and Alzheimer Disease. ACTA ACUST UNITED AC 2020; 4:e2000044. [PMID: 32402127 DOI: 10.1002/adbi.202000044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 12/31/2022]
Abstract
Aging of the circulatory system correlates with the pathogenesis of a large spectrum of diseases. However, it is largely unknown which factors drive the age-dependent or pathological decline of the vasculature and how vascular defects relate to tissue aging. The goal of the study is to design a multianalytical approach to identify how the cellular microenvironment (i.e., fibroblasts) and serum from healthy donors of different ages or Alzheimer disease (AD) patients can modulate the functionality of organ-specific vascular endothelial cells (VECs). Long-living human microvascular networks embedding VECs and fibroblasts from skin biopsies are generated. RNA-seq, secretome analyses, and microfluidic assays demonstrate that fibroblasts from young donors restore the functionality of aged endothelial cells, an effect also achieved by serum from young donors. New biomarkers of vascular aging are validated in human biopsies and it is shown that young serum induces angiopoietin-like-4, which can restore compromised vascular barriers. This strategy is then employed to characterize transcriptional/functional changes induced on the blood-brain barrier by AD serum, demonstrating the importance of PTP4A3 in the regulation of permeability. Features of vascular degeneration during aging and AD are recapitulated, and a tool to identify novel biomarkers that can be exploited to develop future therapeutics modulating vascular function is established.
Collapse
Affiliation(s)
- Simone Bersini
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.,Paul F. Glenn Center for Biology of Aging Research at The Salk Institute, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Rafael Arrojo E Drigo
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core (IGC), The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core (IGC), The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Martin W Hetzer
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
29
|
Li G, Wang X, Yang H, Zhang P, Wu F, Li Y, Zhou Y, Zhang X, Ma H, Zhang W, Li J. α-Linolenic acid but not linolenic acid protects against hypertension: critical role of SIRT3 and autophagic flux. Cell Death Dis 2020; 11:83. [PMID: 32015327 PMCID: PMC6997421 DOI: 10.1038/s41419-020-2277-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
Although dietary α-linolenic acid (ALA) or linolenic acid (LA) intake was reported to be epidemiologically associated with a lower prevalence of hypertension, recent clinical trials have yielded conflicting results. Comparable experimental evidence for the roles of these two different fatty acids is still lacking and the underlying mechanisms need to be further elucidated. Our data showed that ALA but not LA supplementation alleviated systolic blood pressure elevation and improved ACh-induced, endothelium-dependent vasodilation in both spontaneously hypertensive rats (SHRs) and AngII-induced hypertensive mice. In addition, SHRs displayed reduced vascular Sirtuin 3 (SIRT3) expression, subsequent superoxide dismutase 2 (SOD2) hyperacetylation and mitochondrial ROS overproduction, all of which were ameliorated by ALA but not LA supplementation. In primary cultured endothelial cells, ALA treatment directly inhibited SIRT3 reduction, SOD2 hyperacetylation, mitochondrial ROS overproduction and alleviated autophagic flux impairment induced by AngII plus TNFα treatment. However, these beneficial effects of ALA were completely blocked by silencing SIRT3. Restoration of autophagic flux by rapamycin also inhibited mitochondrial ROS overproduction in endothelial cells exposed to AngII plus TNFα. More interestingly, SIRT3 KO mice developed severe hypertension in response to a low dose of AngII infusion, while ALA supplementation lost its anti-hypertensive and endothelium-protective effects on these mice. Our findings suggest that ALA but not LA supplementation improves endothelial dysfunction and diminishes experimental hypertension by rescuing SIRT3 impairment to restore autophagic flux and mitochondrial redox balance in endothelial cells.
Collapse
Affiliation(s)
- Guohua Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinpei Wang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Pengfei Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Fangqin Wu
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yunchu Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yingjie Zhou
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Science, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jia Li
- School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
30
|
Carbone F, Montecucco F, Sahebkar A. Editorial commentary: Promising findings on the role of endothelin-1 and related peptides in primary cardiovascular prevention. Trends Cardiovasc Med 2020; 30:9-10. [PMID: 30808552 DOI: 10.1016/j.tcm.2019.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 11/22/2022]
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino, Genoa, 10 Largo Benzi, 16132, Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Abdelkarim D, Zhao Y, Turner MP, Sivakolundu DK, Lu H, Rypma B. A neural-vascular complex of age-related changes in the human brain: Anatomy, physiology, and implications for neurocognitive aging. Neurosci Biobehav Rev 2019; 107:927-944. [DOI: 10.1016/j.neubiorev.2019.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 01/09/2023]
|
32
|
Diaz-Cañestro C, Reiner MF, Bonetti NR, Liberale L, Merlini M, Wüst P, Amstalden H, Briand-Schumacher S, Semerano A, Giacalone G, Sessa M, Beer JH, Akhmedov A, Lüscher TF, Camici GG. AP-1 (Activated Protein-1) Transcription Factor JunD Regulates Ischemia/Reperfusion Brain Damage via IL-1β (Interleukin-1β). Stroke 2019; 50:469-477. [PMID: 30626291 DOI: 10.1161/strokeaha.118.023739] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background and Purpose- Inflammation is a major pathogenic component of ischemia/reperfusion brain injury, and as such, interventions aimed at inhibiting inflammatory mediators promise to be effective strategies in stroke therapy. JunD-a member of the AP-1 (activated protein-1) family of transcription factors-was recently shown to regulate inflammation by targeting IL (interleukin)-1β synthesis and macrophage activation. The purpose of the present study was to assess the role of JunD in ischemia/reperfusion-induced brain injury. Methods- WT (wild type) mice randomly treated with either JunD or scramble (control) siRNA were subjected to 45 minutes of transient middle cerebral artery occlusion followed by 24 hours of reperfusion. Stroke size, neurological deficit, plasma/brain cytokines, and oxidative stress determined by 4-hydroxynonenal immunofluorescence staining were evaluated 24 hours after reperfusion. Additionally, the role of IL-1β was investigated by treating JunD siRNA mice with an anti-IL-1β monoclonal antibody on reperfusion. Finally, JunD expression was assessed in peripheral blood monocytes isolated from patients with acute ischemic stroke. Results- In vivo JunD knockdown resulted in increased stroke size, reduced neurological function, and increased systemic inflammation, as confirmed by higher neutrophil count and lymphopenia. Brain tissue IL-1β levels were augmented in JunD siRNA mice as compared with scramble siRNA, whereas no difference was detected in IL-6, TNF-α (tumor necrosis factor-α), and 4-hydroxynonenal levels. The deleterious effects of silencing of JunD were rescued by treating mice with an anti-IL-1β antibody. In addition, JunD expression was decreased in peripheral blood monocytes of patients with acute ischemic stroke at 6 and 24 hours after onset of stroke symptoms compared with sex- and age-matched healthy controls. Conclusions- JunD blunts ischemia/reperfusion-induced brain injury via suppression of IL-1β.
Collapse
Affiliation(s)
- Candela Diaz-Cañestro
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Martin F Reiner
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland (M.F.R., J.H.B.)
| | - Nicole R Bonetti
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Luca Liberale
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Italy (L.L.)
| | - Mario Merlini
- Gladstone Institute of Neurological Disease, University of California, San Francisco (M.M.)
| | - Patricia Wüst
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Heidi Amstalden
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Sylvie Briand-Schumacher
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Aurora Semerano
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy (A.S., G.G.)
| | - Giacomo Giacalone
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy (A.S., G.G.)
| | - Maria Sessa
- SC Neurologia, Dipartimento Interaziendale Neuroscienze Cremona-Mantova, Azienda Socio-Sanitaria Territoriale (ASST) di cremona, Cremona, Italy (M.S.)
| | - Jürg H Beer
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Department of Internal Medicine, Cantonal Hospital Baden, Switzerland (M.F.R., J.H.B.)
| | - Alexander Akhmedov
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.)
| | - Thomas F Lüscher
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom (T.F.L.)
| | - Giovanni G Camici
- From the Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland (C.D.-C., M.F.R., N.R.B., L.L., P.W., H.A., S.B.-S., J.H.B., A.A., T.F.L., G.G.C.).,Zurich Neuroscience Center, University of Zurich, Switzerland (G.G.C.)
| |
Collapse
|
33
|
Breau M, Houssaini A, Lipskaia L, Abid S, Born E, Marcos E, Czibik G, Attwe A, Beaulieu D, Palazzo A, Flaman JM, Bourachot B, Collin G, Tran Van Nhieu J, Bernard D, Mechta-Grigoriou F, Adnot S. The antioxidant N-acetylcysteine protects from lung emphysema but induces lung adenocarcinoma in mice. JCI Insight 2019; 4:127647. [PMID: 31578304 DOI: 10.1172/jci.insight.127647] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/31/2019] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress is a major contributor to chronic lung diseases. Antioxidants such as N-acetylcysteine (NAC) are broadly viewed as protective molecules that prevent the mutagenic effects of reactive oxygen species. Antioxidants may, however, increase the risk of some forms of cancer and accelerate lung cancer progression in murine models. Here, we investigated chronic NAC treatment in aging mice displaying lung oxidative stress and cell senescence due to inactivation of the transcription factor JunD, which is downregulated in diseased human lungs. NAC treatment decreased lung oxidative damage and cell senescence and protected from lung emphysema but concomitantly induced the development of lung adenocarcinoma in 50% of JunD-deficient mice and 10% of aged control mice. This finding constitutes the first evidence to our knowledge of a carcinogenic effect of antioxidant therapy in the lungs of aged mice with chronic lung oxidative stress and warrants the utmost caution when considering the therapeutic use of antioxidants.
Collapse
Affiliation(s)
- Marielle Breau
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Amal Houssaini
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Larissa Lipskaia
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Shariq Abid
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Emmanuelle Born
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Elisabeth Marcos
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Gabor Czibik
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Aya Attwe
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Delphine Beaulieu
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Alberta Palazzo
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Lyon, France
| | - Jean-Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Lyon, France
| | - Brigitte Bourachot
- Stress and Cancer Laboratory, Equipe Labelisée LNCC, Institut Curie, INSERM U830, Paris, France
| | - Guillaume Collin
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Lyon, France
| | | | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Lyon, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Equipe Labelisée LNCC, Institut Curie, INSERM U830, Paris, France
| | - Serge Adnot
- INSERM U955, Département de Physiologie-Explorations Fonctionnelles, and DHU A-TVB Hôpital Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
34
|
Liberale L, Camici GG. Longevity-associated variant BPIFB4 gene transfer to recapitulate healthy ageing in patients at risk: is the future around the corner? Eur Heart J 2019; 41:2498-2500. [DOI: 10.1093/eurheartj/ehz522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Good AL, Cannon CE, Haemmerle MW, Yang J, Stanescu DE, Doliba NM, Birnbaum MJ, Stoffers DA. JUND regulates pancreatic β cell survival during metabolic stress. Mol Metab 2019; 25:95-106. [PMID: 31023625 PMCID: PMC6600134 DOI: 10.1016/j.molmet.2019.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/01/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE In type 2 diabetes (T2D), oxidative stress contributes to the dysfunction and loss of pancreatic β cells. A highly conserved feature of the cellular response to stress is the regulation of mRNA translation; however, the genes regulated at the level of translation are often overlooked due to the convenience of RNA sequencing technologies. Our goal is to investigate translational regulation in β cells as a means to uncover novel factors and pathways pertinent to cellular adaptation and survival during T2D-associated conditions. METHODS Translating ribosome affinity purification (TRAP) followed by RNA-seq or RT-qPCR was used to identify changes in the ribosome occupancy of mRNAs in Min6 cells. Gene depletion studies used lentiviral delivery of shRNAs to primary mouse islets or CRISPR-Cas9 to Min6 cells. Oxidative stress and apoptosis were measured in primary islets using cell-permeable dyes with fluorescence readouts of oxidation and activated cleaved caspase-3 and-7, respectively. Gene expression was assessed by RNA-seq, RT-qPCR, and western blot. ChIP-qPCR was used to determine chromatin enrichment. RESULTS TRAP-seq in a PDX1-deficiency model of β cell dysfunction uncovered a cohort of genes regulated at the level of mRNA translation, including the transcription factor JUND. Using a panel of diabetes-associated stressors, JUND was found to be upregulated in mouse islets cultured with high concentrations of glucose and free fatty acid, but not after treatment with hydrogen peroxide or thapsigargin. This induction of JUND could be attributed to increased mRNA translation. JUND was also upregulated in islets from diabetic db/db mice and in human islets treated with high glucose and free fatty acid. Depletion of JUND in primary islets reduced oxidative stress and apoptosis in β cells during metabolic stress. Transcriptome assessment identified a cohort of genes, including pro-oxidant and pro-inflammatory genes, regulated by JUND that are commonly dysregulated in models of β cell dysfunction, consistent with a maladaptive role for JUND in islets. CONCLUSIONS A translation-centric approach uncovered JUND as a stress-responsive factor in β cells that contributes to redox imbalance and apoptosis during pathophysiologically relevant stress.
Collapse
Affiliation(s)
- Austin L Good
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Corey E Cannon
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew W Haemmerle
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Juxiang Yang
- Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Diana E Stanescu
- Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Nicolai M Doliba
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Doris A Stoffers
- Institute for Diabetes, Obesity, and Metabolism and the Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
36
|
Shimizu Y, Yamanashi H, Kitamura M, Furugen R, Iwasaki T, Fukuda H, Hayashida H, Kawasaki K, Kiyoura K, Kawashiri SY, Saito T, Kawakami A, Maeda T. Association between human T cell leukemia virus 1 (HTLV-1) infection and advanced periodontitis in relation to hematopoietic activity among elderly participants: a cross-sectional study. Environ Health Prev Med 2019; 24:42. [PMID: 31182010 PMCID: PMC6558682 DOI: 10.1186/s12199-019-0796-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We reported that human T cell leukemia virus 1 (HTLV-1) infection is positively associated with atherosclerosis. Recent evidence has revealed a close association of periodontitis with atherosclerosis, endothelial dysfunction, and disruption of the microcirculation. However, the association between HTLV-1 and advanced periodontitis has not been investigated to date. Since hematopoietic activity is closely linked to endothelial maintenance activity and is known to decline with age, we hypothesized that the state of hematopoietic activity influenced the association between HTLV-1 and advanced periodontitis in elderly participants. METHODS A cross-sectional study was performed including 822 elderly participants aged 60-99 years who participated in a dental health check-up. Advanced periodontitis was defined as a periodontal pocket ≥ 6.0 mm. Participants were classified as having low or high hematopoietic activity according to the median values of reticulocytes. RESULTS HTLV-1 infection was positively related to advanced periodontitis among participants with lower hematopoietic activity (lower reticulocyte count), but not among participants with higher hematopoietic activity (higher reticulocyte count). The adjusted odds ratio (95% confidence interval) considering potential confounding factors was 1.92 (1.05-3.49) for participants with a lower reticulocyte count and 0.69 (0.35-1.36) for participants with a higher reticulocyte count. CONCLUSIONS Among elderly participants, the association between HTLV-1 infection and advanced periodontitis is influenced by hematopoietic activity. Since hematopoietic activity is associated with endothelial maintenance, these findings provide an efficient tool for clarifying the underlying mechanism of the progression of periodontitis among elderly participants.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523 Japan
- Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Diseases Prevention, Osaka, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Masayasu Kitamura
- Department of Oral Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Reiko Furugen
- Department of Oral Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Iwasaki
- Department of Community Oral Health, School of Dentistry, Asahi University, Gifu, Japan
| | - Hideki Fukuda
- Department of Oral Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hideaki Hayashida
- Department of Oral Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koji Kawasaki
- Community Medical Network Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kairi Kiyoura
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523 Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523 Japan
| | - Toshiyuki Saito
- Department of Oral Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523 Japan
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
37
|
Li J, Zhong Z, Yuan J, Chen X, Huang Z, Wu Z. Resveratrol improves endothelial dysfunction and attenuates atherogenesis in apolipoprotein E-deficient mice. J Nutr Biochem 2019; 67:63-71. [DOI: 10.1016/j.jnutbio.2019.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 11/29/2022]
|
38
|
Dai J, Liu R, Zhao J, Zhang A. Sulfur dioxide improves endothelial dysfunction by downregulating the angiotensin II/AT 1R pathway in D-galactose-induced aging rats. J Renin Angiotensin Aldosterone Syst 2018; 19:1470320318778898. [PMID: 29848151 PMCID: PMC5985551 DOI: 10.1177/1470320318778898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to investigate the protective effects of sulfur dioxide (SO2) on the endothelial function of the aorta in D-galactose (D-gal)-induced aging rats. Sprague Dawley rats were randomized into a D-gal group, a D-gal + SO2 group and a control group, then injected with D-gal, D-gal + SO2 donor or equivalent volumes of saline, respectively, for 8 consecutive weeks. After 8 weeks, the mean arterial pressure was significantly increased in the D-gal group, but was lowered by SO2. SO2 significantly ameliorated the endothelial dysfunction induced by D-gal treatment. The vasorelaxant effect of SO2 was associated with the elevated nitric oxide levels and upregulated phosphorylation of endothelial nitric oxide synthase. In the D-gal group, the concentration of angiotensin II in the plasma was significantly increased, but was decreased by SO2. Moreover, levels of vascular tissue hydrogen peroxide (H2O2) and malondialdehyde were significantly lower in SO2-treated groups than those in the D-gal group. Western blot analysis showed that the expressions of oxidative stress-related proteins (the angiotensin II type 1 receptor (AT1R), and nicotinamide adenine dinucleotide phosphate oxidase subunits) were increased in the D-gal group, while they were decreased after treatment with SO2. In conclusion, SO2 attenuated endothelial dysfunction in association with the inhibition of oxidative stress injury and the downregulation of the angiotensin II/AT1R pathway in D-gal-induced aging rats.
Collapse
Affiliation(s)
- Jing Dai
- 1 Department of Clinical Diagnostics, Hebei Medical University, China
| | - Rui Liu
- 2 Department of Thoracic Surgery, Suining Central Hospital, China
| | - Jinjie Zhao
- 3 Department of Cardiovascular Surgery, Suining Central Hospital, China
| | - Aijie Zhang
- 4 Basic Laboratory, Suining Central Hospital, China
| |
Collapse
|
39
|
Guzik TJ, Cosentino F. Epigenetics and Immunometabolism in Diabetes and Aging. Antioxid Redox Signal 2018; 29:257-274. [PMID: 28891325 PMCID: PMC6012980 DOI: 10.1089/ars.2017.7299] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 09/04/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE A strong relationship between hyperglycemia, impaired insulin pathway, and cardiovascular disease in type 2 diabetes (T2D) is linked to oxidative stress and inflammation. Immunometabolic pathways link these pathogenic processes and pose important potential therapeutic targets. Recent Advances: The link between immunity and metabolism is bidirectional and includes the role of inflammation in the pathogenesis of metabolic disorders such as T2D, obesity, metabolic syndrome, and hypertension and the role of metabolic factors in regulation of immune cell functions. Low-grade inflammation, oxidative stress, balance between superoxide and nitric oxide, and the infiltration of macrophages, T cells, and B cells in insulin-sensitive tissues lead to metabolic impairment and accelerated aging. CRITICAL ISSUES Inflammatory infiltrate and altered immune cell phenotype precede development of metabolic disorders. Inflammatory changes are tightly linked to alterations in metabolic status and energy expenditure and are controlled by epigenetic mechanisms. FUTURE DIRECTIONS A better comprehension of these mechanistic insights is of utmost importance to identify novel molecular targets. In this study, we describe a complex scenario of epigenetic changes and immunometabolism linking to diabetes and aging-associated vascular disease. Antioxid. Redox Signal. 29, 257-274.
Collapse
Affiliation(s)
- Tomasz J. Guzik
- BHF Centre for Research Excellence, Institute of Cardiovascular and Medical Research (ICAMS), University of Glasgow, Glasgow, United Kingdom
- Department of Internal and Agricultural Medicine, Laboratory of Translational Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Madhavan MV, Gersh BJ, Alexander KP, Granger CB, Stone GW. Coronary Artery Disease in Patients ≥80 Years of Age. J Am Coll Cardiol 2018; 71:2015-2040. [DOI: 10.1016/j.jacc.2017.12.068] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/14/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
|
41
|
Costantino S, Camici GG, Mohammed SA, Volpe M, Lüscher TF, Paneni F. Epigenetics and cardiovascular regenerative medicine in the elderly. Int J Cardiol 2018; 250:207-214. [DOI: 10.1016/j.ijcard.2017.09.188] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022]
|
42
|
Enge M, Arda HE, Mignardi M, Beausang J, Bottino R, Kim SK, Quake SR. Single-Cell Analysis of Human Pancreas Reveals Transcriptional Signatures of Aging and Somatic Mutation Patterns. Cell 2017; 171:321-330.e14. [PMID: 28965763 DOI: 10.1016/j.cell.2017.09.004] [Citation(s) in RCA: 348] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/02/2017] [Accepted: 08/30/2017] [Indexed: 12/20/2022]
Abstract
As organisms age, cells accumulate genetic and epigenetic errors that eventually lead to impaired organ function or catastrophic transformation such as cancer. Because aging reflects a stochastic process of increasing disorder, cells in an organ will be individually affected in different ways, thus rendering bulk analyses of postmitotic adult cells difficult to interpret. Here, we directly measure the effects of aging in human tissue by performing single-cell transcriptome analysis of 2,544 human pancreas cells from eight donors spanning six decades of life. We find that islet endocrine cells from older donors display increased levels of transcriptional noise and potential fate drift. By determining the mutational history of individual cells, we uncover a novel mutational signature in healthy aging endocrine cells. Our results demonstrate the feasibility of using single-cell RNA sequencing (RNA-seq) data from primary cells to derive insights into genetic and transcriptional processes that operate on aging human tissue.
Collapse
Affiliation(s)
- Martin Enge
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - H Efsun Arda
- Department of Developmental Biology, Stanford University School of Medicine, CA 94305, USA
| | - Marco Mignardi
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305, USA; Department of Information Technology, Uppsala University, Sweden and SciLifeLab, Uppsala, Sweden SE-751 05
| | - John Beausang
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, CA 94305, USA
| | - Stephen R Quake
- Department of Bioengineering and Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Institute of Cellular Therapeutics, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA.
| |
Collapse
|
43
|
Kidney involvement in psoriasis: a case-control study from China. Int Urol Nephrol 2017; 49:1999-2003. [PMID: 28939941 DOI: 10.1007/s11255-017-1692-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/29/2017] [Indexed: 02/08/2023]
Abstract
PURPOSE Kidney involvement secondary to psoriasis is still a controversial issue. In this study, we aimed to evaluate the prevalence of urinary abnormalities in psoriasis patients and to find out whether the abnormality is related to the severity of psoriasis. METHODS Ninety-seven psoriasis patients (62 females, 35 males, mean age 35.74 ± 13.45 years) and ninety-six age- and gender-matched control subjects (58 males, 38 females, mean age 35.82 ± 13.48 years) without hypertension or diabetes were enrolled in this study. Psoriasis area and severity index (PASI) was used to assess the severity of psoriasis. Twenty-four-hour proteinuria, albuminuria, RBP, and NAG were measured in all patients and controls. Pathologic proteinuria was defined as the total protein excretion of more than 0.4 g/24 h, as measured by the turbidimetric assay. Pathologic albuminuria was defined as albumin excretion of more than 17 mg/24 h. Pathologic NAG and RBP were defined as the excretion of more than 16.5 u/g cr and 0.5 mg/L, respectively. RESULTS Increased 24-h microalbuminuria (11.53 ± 7.29 vs. 9.79 ± 3.72, P = 0.039) and 24-h proteinuria (0.24 ± 0.21 vs. 0.18 ± 0.09, P = 0.002) were found in patients with psoriasis compared with controls. Patients with psoriasis had an increased prevalence of pathological albuminuria (15.46 vs. 5.21%, P = 0.019), NAG (10.31 vs. 3.13%, P = 0.046), and RBP (9.28 vs. 2.08%, P = 0.031) compared with controls. PASI scores in patients with psoriasis correlated positively with 24-h albuminuria (χ 2 = 10.75, P = 0.005). CONCLUSIONS The prevalence of abnormal urinalysis was more common in patients with psoriasis than in controls. The positive correlation between the prevalence of pathological albuminuria and psoriasis severity may indicate a subclinical renal dysfunction in patients with psoriasis.
Collapse
|
44
|
Abstract
Epidemiological studies have shown that ageing is a major non-reversible risk factor for cardiovascular disease. Vascular ageing starts early in life and is characterized by a gradual change of vascular structure and function resulting in increased arterial stiffening. At the present review we discuss the role of the most important molecular pathways involved in vascular ageing, their association with arterial stiffening and possible novel therapeutic targets that may delay this otherwise irreversible degenerating process. Specifically, we discuss the role of oxidative stress, telomere shortening, and ubiquitin proteasome system in endothelial cell senescence and dysfunction in vascular inflammation and in arterial stiffening. Further, we summarize the most important molecular mechanisms regulating vascular ageing including sirtuin 1, telomerase, klotho, JunD, and amyloid beta 1-40 peptide.
Collapse
Affiliation(s)
- Ageliki Laina
- Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece
| | - Konstantinos Stellos
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany; Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Frankfurt, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislaufforschung; DZHK), Rhein-Main Partner Site, Frankfurt, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece.
| |
Collapse
|
45
|
Forte M, Nocella C, De Falco E, Palmerio S, Schirone L, Valenti V, Frati G, Carnevale R, Sciarretta S. The Pathophysiological Role of NOX2 in Hypertension and Organ Damage. High Blood Press Cardiovasc Prev 2017; 23:355-364. [PMID: 27915400 DOI: 10.1007/s40292-016-0175-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
NADPH oxidases (NOXs) represent one of the major sources of reactive oxygen species in the vascular district. Reactive oxygen species are responsible for vascular damage that leads to several cardiovascular pathological conditions. Among NOX isoforms, NOX2 is widely expressed in many cells types, such as cardiomyocytes, endothelial cells, and vascular smooth muscle cells, confirming its pivotal role in vascular pathophysiology. Studies in mice models with systemic deletion of NOX2, as well as in transgenic mice overexpressing NOX2, have demonstrated the undeniable involvement of NOX2 in the development of hypertension, atherosclerosis, diabetes mellitus, cardiac hypertrophy, platelet aggregation, and aging. Of note, the inhibition of NOX2 has been found to be protective for cardiovascular homeostasis. Here, we review the evidence demonstrating that the modulation of NOX2 activity is able to improve vascular physiology, suggesting that NOX2 may be a potential target for therapeutic applications.
Collapse
Affiliation(s)
- Maurizio Forte
- Department of Angiocardioneurology, IRCCS Neuromed, Pozzilli, 86077, Italy
| | - Cristina Nocella
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy
| | - Elena De Falco
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy
| | - Silvia Palmerio
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy
| | - Valentina Valenti
- Department of Imaging, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Giacomo Frati
- Department of Angiocardioneurology, IRCCS Neuromed, Pozzilli, 86077, Italy.,Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy
| | - Roberto Carnevale
- Department of Angiocardioneurology, IRCCS Neuromed, Pozzilli, 86077, Italy
| | - Sebastiano Sciarretta
- Department of Angiocardioneurology, IRCCS Neuromed, Pozzilli, 86077, Italy. .,Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 79 Corso della Repubblica, 04100, Latina, Italy.
| |
Collapse
|
46
|
The Aging Cardiovascular System. J Am Coll Cardiol 2017; 69:1952-1967. [DOI: 10.1016/j.jacc.2017.01.064] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/31/2022]
|
47
|
DNA damage-dependent mechanisms of ageing and disease in the macro- and microvasculature. Eur J Pharmacol 2017; 816:116-128. [PMID: 28347738 DOI: 10.1016/j.ejphar.2017.03.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/07/2017] [Accepted: 03/23/2017] [Indexed: 12/15/2022]
Abstract
A decline in the function of the macro- and micro-vasculature occurs with ageing. DNA damage also accumulates with ageing, and thus DNA damage and repair have important roles in physiological ageing. Considerable evidence also supports a crucial role for DNA damage in the development and progression of macrovascular disease such as atherosclerosis. These findings support the concept that prolonged exposure to risk factors is a major stimulus for DNA damage within the vasculature, in part via the generation of reactive oxygen species. Genomic instability can directly affect vascular cellular function, leading to cell cycle arrest, apoptosis and premature vascular cell senescence. In contrast, the study of age-related impaired function and DNA damage mechanisms in the microvasculature is limited, although ageing is associated with microvessel endothelial dysfunction. This review examines current knowledge on the role of DNA damage and DNA repair systems in macrovascular disease such as atherosclerosis and microvascular disease. We also discuss the cellular responses to DNA damage to identify possible strategies for prevention and treatment.
Collapse
|
48
|
Li Y, Chen F, Deng L, Lin K, Shi X, Zhaoliang S, Wang Y. Febuxostat attenuates paroxysmal atrial fibrillation-induced regional endothelial dysfunction. Thromb Res 2017; 149:17-24. [DOI: 10.1016/j.thromres.2016.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 11/05/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
|
49
|
JunD/AP-1 Antagonizes the Induction of DAPK1 To Promote the Survival of v-Src-Transformed Cells. J Virol 2016; 91:JVI.01925-16. [PMID: 27795443 DOI: 10.1128/jvi.01925-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/07/2016] [Indexed: 01/01/2023] Open
Abstract
The increase in AP-1 activity is a hallmark of cell transformation by tyrosine kinases. Previously, we reported that blocking AP-1 using the c-Jun dominant negative mutant TAM67 induced senescence, adipogenesis, or apoptosis in v-Src-transformed chicken embryo fibroblasts (CEFs) whereas inhibition of JunD by short hairpin RNA (shRNA) specifically induced apoptosis. To investigate the role of AP-1 in Src-mediated transformation, we undertook a gene profiling study to characterize the transcriptomes of v-Src-transformed CEFs expressing either TAM67 or the JunD shRNA. Our study revealed a cluster of 18 probe sets upregulated exclusively in response to AP-1/JunD impairment and v-Src transformation. Four of these probe sets correspond to genes involved in the interferon pathway. One gene in particular, death-associated protein kinase 1 (DAPK1), is a C/EBPβ-regulated mediator of apoptosis in gamma interferon (IFN-γ)-induced cell death. Here, we show that inhibition of DAPK1 abrogates cell death in v-Src-transformed cells expressing the JunD shRNA. Chromatin immunoprecipitation data indicated that C/EBPβ was recruited to the DAPK1 promoter while the expression of a dominant negative mutant of C/EBPβ abrogated the induction of DAPK1 in response to the inhibition of AP-1. In contrast, as determined by chromatin immunoprecipitation (ChIP) assays, JunD was not detected on the DAPK1 promoter under any conditions, suggesting that JunD promotes survival by indirectly antagonizing the expression of DAPK1 in v-Src transformed cells. IMPORTANCE Transformation by the v-Src oncoprotein causes extensive changes in gene expression in primary cells such as chicken embryo fibroblasts. These changes, determining the properties of transformed cells, are controlled in part at the transcriptional level. Much attention has been devoted to transcription factors such as AP-1 and NF-κB and the control of genes associated with a more aggressive phenotype. In this report, we describe a novel mechanism of action determined by the JunD component of AP-1, a factor enhancing cell survival in v-Src-transformed cells. We show that the loss of JunD results in the aberrant activation of a genetic program leading to cell death. This program requires the activation of the tumor suppressor death-associated protein kinase 1 (DAPK1). Since DAPK1 is phosphorylated and inhibited by v-Src, these results highlight the importance of this kinase and the multiple mechanisms controlled by v-Src to antagonize the tumor suppressor function of DAPK1.
Collapse
|
50
|
Antiperoxynitrite Treatment Ameliorates Vasorelaxation of Resistance Arteries in Aging Rats: Involvement With Protection of Circulating Endothelial Progenitor Cells. J Cardiovasc Pharmacol 2016; 68:334-341. [DOI: 10.1097/fjc.0000000000000420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|