1
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-Specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed a Western Diet. Arterioscler Thromb Vasc Biol 2025; 45:74-89. [PMID: 39569521 DOI: 10.1161/atvbaha.124.321366] [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: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pharmacological inhibition of megalin (also known as LRP2 [low-density lipoprotein receptor-related protein-2]) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. METHODS Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and PTC-LRP2 -/- littermates. To study atherosclerosis, all mice were bred to an LDL (low-density lipoprotein) receptor -/- background and fed a Western diet to induce atherosclerosis. RESULTS PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed a Western diet but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not on hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished ALB (albumin) accumulation in PTCs within 10 days of Western diet feeding. RNA-sequencing analyses demonstrated the upregulation of inflammation-related pathways in the kidney. CONCLUSIONS PTC-specific megalin deletion does not affect atherosclerosis but leads to tubulointerstitial nephritis in mice fed a Western diet, with severe pathologies in male mice.
Collapse
MESH Headings
- Animals
- Low Density Lipoprotein Receptor-Related Protein-2/genetics
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Low Density Lipoprotein Receptor-Related Protein-2/deficiency
- Diet, Western/adverse effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Female
- Male
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/metabolism
- Mice
- Mice, Inbred C57BL
- Hypercholesterolemia/genetics
- Hypercholesterolemia/complications
- Hypercholesterolemia/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Plaque, Atherosclerotic
- CD68 Molecule
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Michael K Franklin
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Jessica J Moorleghen
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Deborah A Howatt
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging (Y.K.)
- Department of Biostatistics (Y.K.)
| | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine (M.Y.)
- WPI Institute for the Advanced Study of Human Biology, Kyoto University, Japan (M.Y.)
| | | | - Ruben M Sandoval
- Department of Medicine, Indiana University, Indianapolis (M.M.M.-I., R.M.S., K.W.D.)
| | - Kenneth W Dunn
- Department of Medicine, Indiana University, Indianapolis (M.M.M.-I., R.M.S., K.W.D.)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| | - Hong S Lu
- Saha Cardiovascular Research Center and Saha Aortic Center (N.A., M.K.F., M.K., L.Z., J.J.M., D.A.H., H.S., A.D., H.S.L.)
- Department of Physiology, University of Kentucky, Lexington (H.S., A.D., H.S.L.)
| |
Collapse
|
2
|
Dehghan A, Ardekani A, Akabri M, Sadeghi Y, Radmard AR, Merat S, Khoshnia M, Sharafkhah M, Khuzani AS, Poustchi H, Malekzadeh R, Molavi Vardanjani H. Carotid intima media thickness and glomerular filtration rate: a baseline analysis of the PolyIran-L trial. J Nephrol 2024:10.1007/s40620-024-02122-3. [PMID: 39521753 DOI: 10.1007/s40620-024-02122-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The relationship between kidney and vascular health is acknowledged, but detailed information is still missing. This study examines the relationship of estimated glomerular filtration rate (eGFR) and carotid intima media thickness, providing insights into the association between atherosclerosis and kidney function. METHODS Participants older than 50 years of age who were part of the PolyIran-L study, a trial nested in the Golestan Cohort Study, were included. The maximal intima media thickness of both common carotid arteries was evaluated using B-mode ultrasonography. Four different cut-off values for abnormal carotid intima media thickness were considered. Correlation of carotid intima media thickness and eGFR was assessed with linear correlation and multivariable binary logistic regression models after adjusting for several confounders. RESULTS In total, 1562 participants (750 females, 48%) were included in this population-based study. Assuming the eGFR < 45 [mL/min/1.73 m2] group as reference in the crude analysis, those with eGFR ≥ 45 and < 60 [mL/min/1.73 m2] showed an association of being less likely to have carotid intima media thickness above the 0.8 cutoff. However, the fully adjusted analysis showed no significant statistical association between carotid intima media thickness and eGFR. CONCLUSION This study did not support the independent association of eGFR and different carotid intima media thickness cutoffs. This pattern may be different in patients with severely decreased eGFR, a subset of cases in which it should be further investigated.
Collapse
Affiliation(s)
- Alireza Dehghan
- MPH Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Medical Imaging Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ardekani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Akabri
- Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Sadeghi
- MPH Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Reza Radmard
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Merat
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Disease Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Khoshnia
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Sharafkhah
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Poustchi
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Disease Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Malekzadeh
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Disease Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Molavi Vardanjani
- MD-MPH Department, School of Medicine, Research Center for Traditional Medicine and History of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Zhao J, Liu H, Chen Q, Xia M, Wan L, Yu W, Liu C, Hao X, Tang C, Chen G, Liu Y, Yuan F, Liu H. Mechanistic study of celastrol-mediated inhibition of proinflammatory activation of macrophages in IgA nephropathy via down-regulating ECM1. Int J Biol Sci 2024; 20:5731-5746. [PMID: 39494325 PMCID: PMC11528456 DOI: 10.7150/ijbs.99738] [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: 06/17/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
Increasing evidence suggests that the mononuclear/macrophage system is vital in amplifying the inflammatory cascade in IgA Nephropathy (IgAN). However, the pathogenic mechanism of macrophages in IgAN and targeted treatment strategies still need to be explored. This study found that botanical triterpene celastrol (CLT) effectively alleviated renal lesions, M1-like macrophage infiltration, inflammatory factors production, and improved renal function in IgAN mice. We found that the renal macrophages of IgAN patients had high expression of ECM1, a crucial molecule involved in macrophage inflammatory polarization, positively correlated with the IgAN clinical severity. In murine macrophage Raw 264.7 cells, CLT inhibited macrophage M1-like polarization and the output of TNF-α and IL-6 by downregulating the ECM1/STAT5 pathway. Mechanistically, molecular docking, CESTA, and immunoprecipitation verified that CLT directly bound to ECM1 and increased the ubiquitination of ECM1. Collectively, these results illustrated that CLT inhibited proinflammatory macrophage in IgAN by directly targeting ECM1 to promote ubiquitination degradation of ECM1. Therefore, this study may provide a theoretical basis for exploring the pathogenesis of IgAN and identifying new perspectives for targeted therapy of IgAN.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fang Yuan
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hong Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Li X, Yao C, Lan DM, Wang Y, Qi SC. Porphyromonas gingivalis Induces Chronic Kidney Disease through Crosstalk between the NF-κB/NLRP3 Pathway and Ferroptosis in GMCs. Curr Med Sci 2024; 44:932-946. [PMID: 39446285 DOI: 10.1007/s11596-024-2923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/16/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Porphyromonas gingivalis (P.gingivalis) is a gram-negative bacterium found in the human oral cavity and is a recognized pathogenic bacterium associated with chronic periodontitis and systemic diseases, including chronic kidney disease (CKD), but the roles and molecular mechanism of P.gingivalis in CKD pathogenesis are unclear. METHODS In this study, an animal model of oral P.gingivalis administration and glomerular mesangial cells (GMCs) cocultured with M1-polarized macrophages and P.gingivalis supernatant were constructed. After seven weeks of P.gingivalis gavaged, peripheral blood was collected to detect the changes in renal function. By collecting the teeth and kidneys of mice, H&E staining and IHC were used to analyze the expression of periodontal inflammatory factors in mice, PAS staining was used to analyze glomerular lesions. The supernatant of macrophages was treated with 5% P.gingivalis supernatant. H&E staining, IHC, Western blot and RT-PCR were applied to analyze renal inflammatory factors, macrophage M1 polarization, NF-κB, NLRP3 and ferroptosis changes in vitro. RESULTS We found that oral P.gingivalis administration induced CKD in mice. P.gingivalis supernatant induced macrophage polarization and inflammatory factor upregulation, which triggered the activation of the NF-κB/NLRP3 pathway and ferroptosis in GMCs. By inhibiting the NF-κB/NLRP3 pathway and ferroptosis in GMCs, cell viability and the inflammatory response were partially alleviated in vitro. CONCLUSION We demonstrated that P.gingivalis induced CKD in mice by triggering crosstalk between the NF κB/NLRP3 pathway and ferroptosis in GMCs. Overall, our study suggested that periodontitis can promote the pathogenesis of CKD in mice, which provides evidence of the importance of periodontitis therapy in the prevention and treatment of CKD. P.gingivalis promotes ferroptosis in kidneys and accelerates the progression of CKD through NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xue Li
- Medical College, Anhui University of Science and Technology, Huainan, 232007, China
- Department of Oral Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology Fudan University, Shanghai, 200002, China
| | - Chao Yao
- Department of Oral Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, China
| | - Dong-Mei Lan
- Department of Oral Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, China
| | - Yan Wang
- Medical College, Anhui University of Science and Technology, Huainan, 232007, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology Fudan University, Shanghai, 200002, China.
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200433, China.
| | - Sheng-Cai Qi
- Medical College, Anhui University of Science and Technology, Huainan, 232007, China.
- Department of Oral Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200001, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology Fudan University, Shanghai, 200002, China.
| |
Collapse
|
5
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed Western Diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.592234. [PMID: 38798535 PMCID: PMC11118422 DOI: 10.1101/2024.05.11.592234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Pharmacological inhibition of megalin (also known as low-density lipoprotein receptor-related protein 2: LRP2) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. Methods Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and -/- littermates. To study atherosclerosis, all mice were bred to an LDL receptor -/- background and fed a Western diet to induce atherosclerosis. Results PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed the Western diet, but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished albumin accumulation in PTCs within 10 days of Western diet feeding. RNA sequencing analyses demonstrated the upregulation of inflammation-related pathways in kidney. Conclusions PTC-specific megalin deletion does not affect atherosclerosis, but leads to tubulointerstitial nephritis in mice fed Western diet, with severe pathologies in male mice.
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | | | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
6
|
Zhu Y, Lai Y, Hu Y, Fu Y, Zhang Z, Lin N, Huang W, Zheng L. The mechanisms underlying acute myocardial infarction in chronic kidney disease patients undergoing hemodialysis. Biomed Pharmacother 2024; 177:117050. [PMID: 38968794 DOI: 10.1016/j.biopha.2024.117050] [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: 04/17/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death in chronic kidney disease (CKD). Hemodialysis is one of the main treatments for patients with end-stage kidney disease. Epidemiological data has shown that acute myocardial infarction (AMI) accounts for the main reason for death in patients with CKD under hemodialysis therapy. Immune dysfunction and changes in metabolism (including a high level of inflammatory cytokines, a disorder of lipid and mineral ion homeostasis, accumulation of uremic toxins et al.) during CKD can deteriorate stability of atherosclerotic plaque and promote vascular calcification, which are exactly the pathophysiological mechanisms underlying the occurrence of AMI. Meanwhile, the hemodialysis itself also has adverse effects on lipoprotein, the immune system and hemodynamics, which contribute to the high incidence of AMI in these patients. This review aims to summarize the mechanisms and further promising methods of prevention and treatment of AMI in CKD patients undergoing hemodialysis, which can provide an excellent paradigm for exploring the crosstalk between the kidney and cardiovascular system.
Collapse
Affiliation(s)
- Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Yuchen Lai
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yuxuan Hu
- Hubei University of Science and Technology, Xianning 437100, China
| | - Yiwen Fu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Zheng Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Nan Lin
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350013, China
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China.
| |
Collapse
|
7
|
Wakamatsu T, Yamamoto S, Yoshida S, Narita I. Indoxyl Sulfate-Induced Macrophage Toxicity and Therapeutic Strategies in Uremic Atherosclerosis. Toxins (Basel) 2024; 16:254. [PMID: 38922148 PMCID: PMC11209365 DOI: 10.3390/toxins16060254] [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: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular disease (CVD) frequently occurs in patients with chronic kidney disease (CKD), particularly those undergoing dialysis. The mechanisms behind this may be related to traditional risk factors and CKD-specific factors that accelerate atherosclerosis and vascular calcification in CKD patients. The accumulation of uremic toxins is a significant factor in CKD-related systemic disorders. Basic research suggests that indoxyl sulfate (IS), a small protein-bound uremic toxin, is associated with macrophage dysfunctions, including increased oxidative stress, exacerbation of chronic inflammation, and abnormalities in lipid metabolism. Strategies to mitigate the toxicity of IS include optimizing gut microbiota, intervening against the abnormality of intracellular signal transduction, and using blood purification therapy with higher efficiency. Further research is needed to examine whether lowering protein-bound uremic toxins through intervention leads to a reduction in CVD in patients with CKD.
Collapse
Affiliation(s)
- Takuya Wakamatsu
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
- Ohgo Clinic, Maebashi 371-0232, Japan
| | - Suguru Yamamoto
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| | - Shiori Yoshida
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| |
Collapse
|
8
|
Ushio Y, Kataoka H, Akagawa H, Sato M, Manabe S, Kawachi K, Makabe S, Akihisa T, Seki M, Teraoka A, Iwasa N, Yoshida R, Tsuchiya K, Nitta K, Hoshino J, Mochizuki T. Factors associated with early-onset intracranial aneurysms in patients with autosomal dominant polycystic kidney disease. J Nephrol 2024; 37:983-992. [PMID: 38315279 DOI: 10.1007/s40620-023-01866-8] [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: 05/22/2023] [Accepted: 12/14/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Recently, the importance of attribute-based medicine has been emphasized. The effects of early-onset intracranial aneurysms on patients can be significant and long-lasting. Herein, we compared the factors associated with intracranial aneurysms in patients with autosomal dominant polycystic kidney disease (ADPKD) according to age categories (≥ 50 years, < 50 years). METHODS We included 519 ADPKD patients, with a median age of 44 years, estimated glomerular filtration rate of 54.5 mL/min/1.73 m2, and total follow-up duration of 3104 patient-years. Logistic regression analyses were performed to determine factors associated with intracranial aneurysms. RESULTS Regarding the presence of intracranial aneurysm, significant interactions were identified between the age category (age ≥ 50 years), female sex (P = 0.0027 for the interaction) and hypertension (P = 0.0074 for the interaction). Female sex and hypertension were associated with intracranial aneurysm risk factors only in patients aged ≥ 50 years. The presence of intracranial aneurysm was significantly associated with chronic kidney disease (CKD) stages 4-5 (odds ratio [OR] = 3.87, P = 0.0007) and family history of intracranial aneurysm or subarachnoid hemorrhage (OR = 2.30, P = 0.0217) in patients aged < 50 years. For patients aged ≥ 50 years, in addition to the abovementioned factors [OR = 2.38, P = 0.0355 for CKD stages 4-5; OR = 3.49, P = 0.0094 for family history of intracranial aneurysm or subarachnoid hemorrhage], female sex (OR = 4.51, P = 0.0005), and hypertension (OR = 5.89, P = 0.0012) were also associated with intracranial aneurysm. CONCLUSION Kidney dysfunction and family history of intracranial aneurysm or subarachnoid hemorrhage are risk factors for early-onset intracranial aneurysm. Patients aged < 50 years with a family history of intracranial aneurysm or subarachnoid hemorrhage or with CKD stages 4-5 may be at an increased risk of early-onset intracranial aneurysm.
Collapse
Affiliation(s)
- Yusuke Ushio
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Hiroyuki Akagawa
- Tokyo Women's Medical University Institute for Integrated Medical Sciences (TIIMS), Tokyo, Japan
| | - Masayo Sato
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Keiko Kawachi
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Shiho Makabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Taro Akihisa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Momoko Seki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Atsuko Teraoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Naomi Iwasa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Rie Yoshida
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Ken Tsuchiya
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Kosaku Nitta
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Toshio Mochizuki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
9
|
Shimizu M, Fujii H, Kono K, Goto S, Watanabe K, Sakamoto K, Nishi S. Clinical Implication of Consistently Strict Phosphate Control for Coronary and Valvular Calcification in Incident Patients Undergoing Hemodialysis. J Atheroscler Thromb 2023; 30:1568-1579. [PMID: 36990726 PMCID: PMC10627770 DOI: 10.5551/jat.64159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
AIMS Serum phosphate control is crucial for the progression of vascular and valvular calcifications. Strict phosphate control is recently suggested; however, there is a lack of convincing evidence. Therefore, we explored the effects of strict phosphate control on vascular and valvular calcifications in incident patients undergoing hemodialysis. METHODS A total of 64 patients undergoing hemodialysis from our previous randomized controlled trial were included in this study. Coronary artery calcification score (CACS) and cardiac valvular calcification score (CVCS) were evaluated using computed tomography and ultrasound cardiography at baseline and 18 months after the initiation of hemodialysis. The absolute changes in CACS (ΔCACS) and CVCS (ΔCVCS) and the percent change in CACS (%ΔCACS) and CVCS (%ΔCVCS) were calculated. Serum phosphate level was measured at 6, 12, and 18 months after the initiation of hemodialysis. Moreover, phosphate control status was evaluated using the area under the curve (AUC) by the amount of time spent with a serum phosphate level of ≥ 4.5 mg/dL and the extent to which this threshold exceeded over the observation period. RESULTS ΔCACS, %ΔCACS, ΔCVCS, and %ΔCVCS were significantly lower in the low AUC group than in the high AUC group. ΔCACS and %ΔCACS were also significantly lower. ΔCVCS and %ΔCVCS tended to be lower in patients whose serum phosphate level never exceeded 4.5 mg/dL than in those whose serum phosphate level continuously exceeded 4.5 mg/dL. AUC significantly correlated with ΔCACS and ΔCVCS. CONCLUSION Consistently strict phosphate control may slow the progression of coronary and valvular calcifications in incident patients undergoing hemodialysis.
Collapse
Affiliation(s)
- Mao Shimizu
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideki Fujii
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keiji Kono
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shunsuke Goto
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kentaro Watanabe
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazuo Sakamoto
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinichi Nishi
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
10
|
Mazzarino M, Cetin E, Bartosova M, Marinovic I, Ipseiz N, Hughes TR, Schmitt CP, Ramji DP, Labéta MO, Raby AC. Therapeutic targeting of chronic kidney disease-associated DAMPs differentially contributing to vascular pathology. Front Immunol 2023; 14:1240679. [PMID: 37849759 PMCID: PMC10577224 DOI: 10.3389/fimmu.2023.1240679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 10/19/2023] Open
Abstract
Chronic Kidney Disease (CKD) is associated with markedly increased cardiovascular (CV) morbidity and mortality. Chronic inflammation, a hallmark of both CKD and CV diseases (CVD), is believed to drive this association. Pro-inflammatory endogenous TLR agonists, Damage-Associated Molecular Patterns (DAMPs), have been found elevated in CKD patients' plasma and suggested to promote CVD, however, confirmation of their involvement, the underlying mechanism(s), the extent to which individual DAMPs contribute to vascular pathology in CKD and the evaluation of potential therapeutic strategies, have remained largely undescribed. A multi-TLR inhibitor, soluble TLR2, abrogated chronic vascular inflammatory responses and the increased aortic atherosclerosis-associated gene expression observed in nephropathic mice, without compromising infection clearance. Mechanistically, we confirmed elevation of 4 TLR DAMPs in CKD patients' plasma, namely Hsp70, Hyaluronic acid, HMGB-1 and Calprotectin, which displayed different abilities to promote key cellular responses associated with vascular inflammation and progression of atherosclerosis in a TLR-dependent manner. These included loss of trans-endothelial resistance, enhanced monocyte migration, increased cytokine production, and foam cell formation by macrophages, the latter via cholesterol efflux inhibition. Calprotectin and Hsp70 most consistently affected these functions. Calprotectin was further elevated in CVD-diagnosed CKD patients and strongly correlated with the predictor of CV events CRP. In nephropathic mice, Calprotectin blockade robustly reduced vascular chronic inflammatory responses and pro-atherosclerotic gene expression in the blood and aorta. Taken together, these findings demonstrated the critical extent to which the DAMP-TLR pathway contributes to vascular inflammatory and atherogenic responses in CKD, revealed the mechanistic contribution of specific DAMPs and described two alternatives therapeutic approaches to reduce chronic vascular inflammation and lower CV pathology in CKD.
Collapse
Affiliation(s)
- Morgane Mazzarino
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Esra Cetin
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Natacha Ipseiz
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Timothy R. Hughes
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Dipak P. Ramji
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mario O. Labéta
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
11
|
Mallén A, Rodriguez-Urquia R, Alvarez R, Dorca-Duch E, Navarro E, Hueso M. Sex Differences in Glomerular Lesions, in Atherosclerosis Progression, and in the Response to Angiotensin-Converting Enzyme Inhibitors in the ApoE -/- Mice Model. Int J Mol Sci 2023; 24:13442. [PMID: 37686247 PMCID: PMC10487579 DOI: 10.3390/ijms241713442] [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: 08/07/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
This study analyzes sex-based differences in renal structure and the response to the Angiotensin-Converting Enzyme (ACE) inhibitor enalapril in a mouse model of atherosclerosis. Eight weeks old ApoE-/- mice received enalapril (5 mg/kg/day, subcutaneous) or PBS (control) for an additional 14 weeks. Each group consisted of six males and six females. Females exhibited elevated LDL-cholesterol levels, while males presented higher creatinine levels and proteinuria. Enalapril effectively reduced blood pressure in both groups, but proteinuria decreased significantly only in females. Plaque size analysis and assessment of kidney inflammation revealed no significant sex-based differences. However, males displayed more severe glomerular injury, with increased mesangial expansion, mesangiolysis, glomerular foam cells, and activated parietal epithelial cells (PECs). Enalapril mitigated mesangial expansion, glomerular inflammation (particularly in the female group), and hypertrophy of the PECs in males. This study demonstrates sex-based differences in the response to enalapril in a mouse model of atherosclerosis. Males exhibited more severe glomerular injury, while enalapril provided renal protection, particularly in females. These findings suggest potential sex-specific considerations for ACE inhibitor therapy in chronic kidney disease and atherosclerosis cardiovascular disease. Further research is needed to elucidate the underlying mechanism behind these observations.
Collapse
Affiliation(s)
- Adrián Mallén
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
| | - Ronny Rodriguez-Urquia
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| | - Rafael Alvarez
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| | - Eduard Dorca-Duch
- Department of Pathology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain;
| | - Estanis Navarro
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
| | - Miguel Hueso
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| |
Collapse
|
12
|
Tanriover B, Stewart D, Kamal L, Saeed M, Cooper M, Foutz J, McGehee H, Gupta G. The Independent Effects of Kidney Length and Vascular Plaque on Ten-Year Outcomes of Extended Criteria Donor Kidney Transplants. Transpl Int 2023; 36:11373. [PMID: 37519905 PMCID: PMC10379651 DOI: 10.3389/ti.2023.11373] [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: 03/19/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023]
Abstract
The independent effects of deceased donor kidney length and vascular plaque on long-term graft survival are not established. Utilizing DonorNet attachments from 4,480 expanded criteria donors (ECD) recovered between 2008 and 2012 in the United States with at least one kidney biopsied and transplanted, we analyzed the relationship between kidney length and vascular plaques and 10-year hazard of all-cause graft failure (ACGF) using causal inference methods in a Cox regression framework. The composite plaque score (range 0-4) and the presence of any plaque (yes, no) was also analyzed. Kidney length was modeled both categorically (<10, 10-12, >12 cm) as well as numerically, using a restricted cubic spline to capture nonlinearity. Effects of a novel composite plaque score 4 vs. 0 (HR 1.08; 95% CI: 0.96, 1.23) and the presence of any vascular plaque (HR 1.08; 95% CI: 0.98, 1.20) were attenuated after adjustment. Likewise, we identified a potential nonlinear relationship between kidney length and the 10-year hazard of ACGF, however the strength of the relationship was attenuated after adjusting for other donor factors. The independent effects of vascular plaque and kidney length on long-term ECD graft survival were found to be minimal and should not play a significant role in utilization.
Collapse
Affiliation(s)
- Bekir Tanriover
- Division of Nephrology, University of Arizona, Tucson, AZ, United States
| | - Darren Stewart
- Department of Surgery, New York University Langone Health, New York City, NY, United States
| | - Layla Kamal
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA, United States
| | - Muhammad Saeed
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA, United States
| | - Matthew Cooper
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Julia Foutz
- United Network for Organ Sharing, Richmond, VA, United States
| | | | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
13
|
Joo YS, Rim TH, Koh HB, Yi J, Kim H, Lee G, Kim YA, Kang SW, Kim SS, Park JT. Non-invasive chronic kidney disease risk stratification tool derived from retina-based deep learning and clinical factors. NPJ Digit Med 2023; 6:114. [PMID: 37330576 DOI: 10.1038/s41746-023-00860-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 06/09/2023] [Indexed: 06/19/2023] Open
Abstract
Despite the importance of preventing chronic kidney disease (CKD), predicting high-risk patients who require active intervention is challenging, especially in people with preserved kidney function. In this study, a predictive risk score for CKD (Reti-CKD score) was derived from a deep learning algorithm using retinal photographs. The performance of the Reti-CKD score was verified using two longitudinal cohorts of the UK Biobank and Korean Diabetic Cohort. Validation was done in people with preserved kidney function, excluding individuals with eGFR <90 mL/min/1.73 m2 or proteinuria at baseline. In the UK Biobank, 720/30,477 (2.4%) participants had CKD events during the 10.8-year follow-up period. In the Korean Diabetic Cohort, 206/5014 (4.1%) had CKD events during the 6.1-year follow-up period. When the validation cohorts were divided into quartiles of Reti-CKD score, the hazard ratios for CKD development were 3.68 (95% Confidence Interval [CI], 2.88-4.41) in the UK Biobank and 9.36 (5.26-16.67) in the Korean Diabetic Cohort in the highest quartile compared to the lowest. The Reti-CKD score, compared to eGFR based methods, showed a superior concordance index for predicting CKD incidence, with a delta of 0.020 (95% CI, 0.011-0.029) in the UK Biobank and 0.024 (95% CI, 0.002-0.046) in the Korean Diabetic Cohort. In people with preserved kidney function, the Reti-CKD score effectively stratifies future CKD risk with greater performance than conventional eGFR-based methods.
Collapse
Affiliation(s)
- Young Su Joo
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea
| | - Tyler Hyungtaek Rim
- Mediwhale Inc, Seoul, Republic of Korea.
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore.
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore.
| | - Hee Byung Koh
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Republic of Korea
- Department of Internal Medicine, International Saint Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Joseph Yi
- Albert Einstein College of Medicine, New York, USA
| | | | | | - Young Ah Kim
- Division of Digital Health, Yonsei University Health System, Seoul, Republic of Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Republic of Korea
| | - Sung Soo Kim
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
14
|
The Development of Dyslipidemia in Chronic Kidney Disease and Associated Cardiovascular Damage, and the Protective Effects of Curcuminoids. Foods 2023; 12:foods12050921. [PMID: 36900438 PMCID: PMC10000737 DOI: 10.3390/foods12050921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Chronic kidney disease (CKD) is a health problem that is constantly growing. This disease presents a diverse symptomatology that implies complex therapeutic management. One of its characteristic symptoms is dyslipidemia, which becomes a risk factor for developing cardiovascular diseases and increases the mortality of CKD patients. Various drugs, particularly those used for dyslipidemia, consumed in the course of CKD lead to side effects that delay the patient's recovery. Therefore, it is necessary to implement new therapies with natural compounds, such as curcuminoids (derived from the Curcuma longa plant), which can cushion the damage caused by the excessive use of medications. This manuscript aims to review the current evidence on the use of curcuminoids on dyslipidemia in CKD and CKD-induced cardiovascular disease (CVD). We first described oxidative stress, inflammation, fibrosis, and metabolic reprogramming as factors that induce dyslipidemia in CKD and their association with CVD development. We proposed the potential use of curcuminoids in CKD and their utilization in clinics to treat CKD-dyslipidemia.
Collapse
|
15
|
Zhong S, Cai Q, Zhong L, Wang Y, Liang F, Deng Z, Li S, Zha D, Qiu W, Wu J. Low-Intensity Focused Ultrasound Ameliorates Ischemic Heart Failure Related to the Cholinergic Anti-Inflammatory Pathway. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:463-475. [PMID: 36444908 DOI: 10.1002/jum.16140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/30/2022] [Accepted: 11/13/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES This study aims to determine the effect of low-intensity focused ultrasound (LIFU) in ischemic heart failure (IHF) and explore the potential neuroimmune mechanism. METHODS Sprague-Dawley rats were subjected to ultrasound (US) with specific parameters, and electrocardiograms were recorded to analyze the effect of LIFU and/or vagal denervation on heart rate. Thereafter, myocardial infarction (MI) was induced by left anterior artery ligation, and LIFU was performed three times a day for 25 days after MI. Echocardiography, Masson staining, and ELISA were used to evaluate the effect of LIFU on the structure and function of the heart. Finally, ELISA, flow cytometry, qRT-PCR, and Western blot analysis were performed to determine the effect of LIFU on the inflammation and the expression of the cholinergic anti-inflammatory pathway (CAP)-related mediators. RESULTS LIFU reduced heart rate in rats (control vs LIFU, P < .01), and vagotomy (VT) eliminated this effect of LIFU on heart rate (VT vs LIFU + VT, P > .01). LIFU-ameliorated IHF in terms of cardiac structure and function (MI vs MI + LIFU, P < .01), but VT abrogated the beneficial effect of LIFU (MI + VT vs MI + LIFU + VT, P > .01). After the treatment of LIFU, decreased levels of inflammatory cytokines, increased proportion of anti-inflammatory macrophages, and increased expression of CAP-related mediators (MI vs MI + LIFU, P < .01). CONCLUSIONS LIFU ameliorates IHF whereas the CAP plays a promising role. LIFU has the potential to be a novel nonpharmacological and noninvasive therapy for the treatment of coronary artery disease and other cardiovascular diseases.
Collapse
Affiliation(s)
- Shenrong Zhong
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology, Meizhou People's Hospital, Meizhou, China
| | - Qianyun Cai
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Longhe Zhong
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuegang Wang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fengchu Liang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe Deng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shasha Li
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Daogang Zha
- Department of General Practice, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weibao Qiu
- Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, Chinese Academy of Sciences, Shenzhen, China
| | - Juefei Wu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
| |
Collapse
|
16
|
Chen A, Lee K, He JC. Treating crescentic glomerulonephritis by targeting macrophages. Kidney Int 2022; 102:1212-1214. [PMID: 36411015 DOI: 10.1016/j.kint.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
Macrophage accumulation in the kidney is associated with the progression of crescentic glomerulonephritis (GN) and is mostly derived from circulating monocytes. FROUNT, a C-C motif chemokine receptor 2 (CCR2)-interacted protein, which is strongly expressed in monocytes/macrophages, enhances macrophage infiltration through CCR2-mediated chemotaxis. In this issue of the journal, Toda et al. reported that disulfiram, an inhibitor of FROUNT, attenuates GN by inhibition of the FROUNT-CCR2 interaction and macrophage migration and activation, suggesting a potential therapeutic role for crescentic GN.
Collapse
Affiliation(s)
- Anqun Chen
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China.
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, Bronx, New York, USA.
| |
Collapse
|
17
|
Lin CW, Armstrong DG, Huang CH, Lin CH, Hung SY, Liu PH, Huang YY. Diabetic foot disease in subjects with End-stage renal Disease: A nationwide study over 14 years highlighting an emerging threat. Diabetes Res Clin Pract 2022; 193:110134. [PMID: 36349589 DOI: 10.1016/j.diabres.2022.110134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
Abstract
AIMS To disclose prevalence, demographic, foot characteristics as well as management and lower-extremity amputations (LEAs) of subjects with end-stage renal disease (ESRD) on diabetic foot diseases (DFDs). METHODS Data were derived from the Taiwan National Health Insurance Research Database between 2004 and 2017. DFDs were defined as ulcers, infections, or severe peripheral arterial diseases (PADs) in patients with type 2 diabetes. Clinical characteristics were analyzed between subjects with and without ESRD. RESULTS Subjects with ESRD have increased impacts on the DFD population either from annual prevalence (2.7 % to 10.42 %, P for trend < 0.001), or proportional representation in LEAs (7.91 % to 26.37 %, P < 0.001) over 14 years. The annual trends for major-LEAs rates have decreased in both subjects with and without ESRD (13.67 % to 5.82 % and 3.48 % to 1.47 %, both P < 0.001). Notably, the concomitant increase of endovascular treatments (EVTs) (7.09 % to 29.41 %, P < 0.001) was associated with the decrease of major-LEAs (P for interaction < 0.001) in subjects with ESRD. CONCLUSIONS As the annual prevalence of subjects with ESRD has increased 3.9-fold over years, they now account for more than 30% of annual major-LEA of the total DFD population. Interdisciplinary team approach and aggressive EVTs might reduce major-LEAs in these patients.
Collapse
Affiliation(s)
- Cheng-Wei Lin
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - David G Armstrong
- Southwestern Academic Limb Salvage Alliance (SALSA), Department of Surgery, Keck School of Medicine of University of Southern California (USC), Los Angeles, USA (D.G.A.)
| | - Chung-Huei Huang
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Chia-Hung Lin
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Shih-Yuan Hung
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Pi-Hua Liu
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan; Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Yu-Yao Huang
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan; College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Department of Medical Nutrition Therapy, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.
| |
Collapse
|
18
|
Poznyak AV, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. Atherosclerosis Specific Features in Chronic Kidney Disease (CKD). Biomedicines 2022; 10:biomedicines10092094. [PMID: 36140195 PMCID: PMC9495595 DOI: 10.3390/biomedicines10092094] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Atherosclerosis is the major cause of cardiovascular disease, leading to a high mortality rate worldwide. Several risk factors are known to favor atherogenesis, among which are high blood pressure, smoking, diabetes mellitus, and others. Chronic kidney disease is another serious health problem associated with significant health care costs, morbidity, and mortality. Chronic kidney disease shares several risk factors with atherosclerosis and cardiovascular diseases, such as hypertension and diabetes mellitus. Additional risk factors for cardiovascular disease development should be considered in patients with chronic kidney disease. Interestingly, patients suffering from chronic kidney disease are more prone to cardiovascular problems than the general population. Moreover, chronic kidney disease is characterized by an increased atherosclerotic burden from the very early stages. The purpose of this review was to summarize data on atherosclerosis in chronic kidney disease, highlighting the specific features of the disease combination.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Andrey G. Kartuesov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Evgeny E. Borisov
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
19
|
Pan X. The Roles of Fatty Acids and Apolipoproteins in the Kidneys. Metabolites 2022; 12:metabo12050462. [PMID: 35629966 PMCID: PMC9145954 DOI: 10.3390/metabo12050462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The kidneys are organs that require energy from the metabolism of fatty acids and glucose; several studies have shown that the kidneys are metabolically active tissues with an estimated energy requirement similar to that of the heart. The kidneys may regulate the normal and pathological function of circulating lipids in the body, and their glomerular filtration barrier prevents large molecules or large lipoprotein particles from being filtered into pre-urine. Given the permeable nature of the kidneys, renal lipid metabolism plays an important role in affecting the rest of the body and the kidneys. Lipid metabolism in the kidneys is important because of the exchange of free fatty acids and apolipoproteins from the peripheral circulation. Apolipoproteins have important roles in the transport and metabolism of lipids within the glomeruli and renal tubules. Indeed, evidence indicates that apolipoproteins have multiple functions in regulating lipid import, transport, synthesis, storage, oxidation and export, and they are important for normal physiological function. Apolipoproteins are also risk factors for several renal diseases; for example, apolipoprotein L polymorphisms induce kidney diseases. Furthermore, renal apolipoprotein gene expression is substantially regulated under various physiological and disease conditions. This review is aimed at describing recent clinical and basic studies on the major roles and functions of apolipoproteins in the kidneys.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA;
- Diabetes and Obesity Research Center, NYU Langone Hospital—Long Island, Mineola, New York, NY 11501, USA
| |
Collapse
|
20
|
Al-rawi KF, Ali HH, Guma MA, Mohammed Aldahham BJ, Tuleab Alaaraji SF, Al-ani O, Tariq Ali A. Relationship Between IL-2, IL-17 Concentrations, and Serum Creatinine Levels in Men with Chronic Kidney Diseases. Rep Biochem Mol Biol 2022; 10:664-674. [PMID: 35291613 PMCID: PMC8903360 DOI: 10.52547/rbmb.10.4.664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/19/2021] [Indexed: 06/14/2023]
Abstract
Background Chronic kidney disease (CKD), is a major public health challenge worldwide. It is more prevalent in developed countries compared with the rest of the world, due to the higher rates of life expectancy and unhealthy lifestyle related factors. This aim of the current study is to evaluate the relationship between interleukins IL-2 and IL-17 concentrations and kidney function markers in men with CKD. Methods Forty-five men with CKD and seventy controls were enrolled in the current study to assess the relationship between interleukin-2 (IL-2), interleukin-17 (IL-17), and CKD parameters. Fasting blood samples were collected from patients with CKD and their controls at same time. Serum IL-2, and IL-17 were measured in patients with CKD and their controls, and then the relationship between these interleukins and serum creatinine, serum urea, serum uric acid and urine albumin were evaluated. Results A significant relationship was detected between IL-2 (p< 0.001), IL-17 (p< 0.001) levels and serum creatinine concentrations. The significant increase of IL-2 and IL-17 levels were also paralleled with a significant increase in serum urea (p< 0.001), and urine albumin (p< 0.001) concentrations respectively. Conclusion IL-2 and IL-17 may play a critical role in the pathophysiology of CKD. The significant increase of IL-2 and IL-17 is associated with significantly high concentrations of creatinine, serum urea and urine albumin suggesting that these interleukins may be used as targets for future biomarkers and molecular therapy. However, due to limited sample size of the current study, larger prospective cohorts are needed to confirm these observations.
Collapse
Affiliation(s)
- Khalid Farouq Al-rawi
- Department of Chemistry, College of Science, University of Anbar, Al-anbar Province, Iraq.
| | - Hameed Hussein Ali
- Department of Chemistry, College of Science, University of Anbar, Al-anbar Province, Iraq.
| | - Manaf Abdulrahman Guma
- Department of Applied Chemistry, College of Applied Sciences, University of Anbar, Al-anbar Province, Iraq.
| | | | | | | | | |
Collapse
|
21
|
El-Senosy FM, Abd El Aziz REM, Kasim SA, Gad LA, Hassan DA, Sabry S, El mancy IM, Shawky TA, Mohamed IGR, Elmonier R, Kotb E, Abdul-Mohymen AM. Serum Endocan Levels and Subclinical Atherosclerosis in Patients with Chronic Kidney and End-Stage Renal Diseases. Int J Clin Pract 2022; 2022:4524637. [PMID: 35936065 PMCID: PMC9300358 DOI: 10.1155/2022/4524637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
RESULTS HD and CKD groups had significantly higher endocan levels when compared with control group (median (IQR): 519.0 (202.3-742.0) versus 409.0 (245.3-505.3) and 273.0 (168.0-395.5) ng/L, respectively). Also, HD patients had significantly higher endocan levels when compared with CKD levels. HD patients had significantly higher carotid intima-media thickness (CIMT) when compared with CKD patients (median (IQR): 0.80 (0.80-0.90) versus 0.75 (0.73-0.75) mm, p < 0.001). HD patients had significantly higher frequency of SCA when compared with CKD patients (46.7% versus 13.3%, p=0.005). Patients with SCA had significantly higher hsCRP (median (IQR): 36.5 (26.8-43.5) versus 24.0 (15.8-29.0) mg/dl) and endocan levels (697.0 (528.3-974.8) versus 222.5 (158.8-565.8) ng/L) when compared with patients without SCA. ROC curve analysis of endocan for identification of SCA in HD patients showed that at a cutoff of 380.5 ng/L, endocan has an AUC of 0.862 with a sensitivity and specificity of 92.9% and 68.7%, respectively. CONCLUSIONS Serum endocan levels are related to SCA in HD patients. In addition, it is associated with the hyperinflammatory state in those patients.
Collapse
Affiliation(s)
- Fatma M. El-Senosy
- Department of Internal Medicine, Faculty of Medicine (Girls) Al-Azhar University, Cairo, Egypt
| | | | - Sammar Ahmed Kasim
- Department of Internal Medicine, Faculty of Medicine (Girls) Al-Azhar University, Cairo, Egypt
| | - Lamia Abdulbary Gad
- Department of Clinical Pathology, Faculty of Medicine (Girls) Al-Azhar University, Cairo, Egypt
| | - Donia Ahmed Hassan
- Department of Clinical Pathology, Faculty of Medicine (Girls) Al-Azhar University, Cairo, Egypt
| | - Seham Sabry
- Department of Internal Medicine, Faculty of Medicine (Girls) Al-Azhar University, Cairo, Egypt
| | | | - Taiseer Ahmed Shawky
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | - Rady Elmonier
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Essam Kotb
- Department of Internal Medicine, Faculty of Medicine, October 6 University, 6th of October, Egypt
| | | |
Collapse
|
22
|
Pro-Inflammatory Serum Amyloid a Stimulates Renal Dysfunction and Enhances Atherosclerosis in Apo E-Deficient Mice. Int J Mol Sci 2021; 22:ijms222212582. [PMID: 34830462 PMCID: PMC8623330 DOI: 10.3390/ijms222212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Acute serum amyloid A (SAA) is an apolipoprotein that mediates pro-inflammatory and pro-atherogenic pathways. SAA-mediated signalling is diverse and includes canonical and acute immunoregulatory pathways in a range of cell types and organs. This study aimed to further elucidate the roles for SAA in the pathogenesis of vascular and renal dysfunction. Two groups of male ApoE-deficient mice were administered SAA (100 µL, 120 µg/mL) or vehicle control (100 µL PBS) and monitored for 4 or 16 weeks after SAA treatment; tissue was harvested for biochemical and histological analyses at each time point. Under these conditions, SAA administration induced crosstalk between NF-κB and Nrf2 transcriptional factors, leading to downstream induction of pro-inflammatory mediators and antioxidant response elements 4 weeks after SAA administration, respectively. SAA treatment stimulated an upregulation of renal IFN-γ with a concomitant increase in renal levels of p38 MAPK and matrix metalloproteinase (MMP) activities, which is linked to tissue fibrosis. In the kidney of SAA-treated mice, the immunolocalisation of inducible nitric oxide synthase (iNOS) was markedly increased, and this was localised to the parietal epithelial cells lining Bowman’s space within glomeruli, which led to progressive renal fibrosis. Assessment of aortic root lesion at the study endpoint revealed accelerated atherosclerosis formation; animals treated with SAA also showed evidence of a thinned fibrous cap as judged by diffuse collagen staining. Together, this suggests that SAA elicits early renal dysfunction through promoting the IFN-γ-iNOS-p38 MAPK axis that manifests as the fibrosis of renal tissue and enhanced cardiovascular disease.
Collapse
|
23
|
Prasad K. Current Status of Primary, Secondary, and Tertiary Prevention of Coronary Artery Disease. Int J Angiol 2021; 30:177-186. [PMID: 34776817 PMCID: PMC8580611 DOI: 10.1055/s-0041-1731273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Fifty percent of all death from cardiovascular diseases is due to coronary artery disease (CAD). This is avoidable if early identification is made. Preventive health care has a major role in the fight against CAD. Atherosclerosis and atherosclerotic plaque rupture are involved in the development of CAD. Modifiable risk factors for CAD are dyslipidemia, diabetes, hypertension, cigarette smoking, obesity, chronic renal disease, chronic infection, high C-reactive protein, and hyperhomocysteinemia. CAD can be prevented by modification of risk factors. This paper defines the primary, secondary, and tertiary prevention of CAD. It discusses the mechanism of risk factor-induced atherosclerosis. This paper describes the CAD risk score and its use in the selection of individuals for primary prevention of CAD. Guidelines for primary, secondary, and tertiary prevention of CAD have been described. Modification of risk factors and use of guidelines for prevention of CAD would prevent, regress, and slow down the progression of CAD, improve the quality of life of patient, and reduce the health care cost.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology (APP), College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
24
|
Features of asymptomatic contralateral limb in patients with chronic limb-threatening ischemia. J Cardiol 2021; 79:153-160. [PMID: 34470712 DOI: 10.1016/j.jjcc.2021.08.021] [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: 04/23/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The current study aimed to reveal clinical features and prognosis of asymptomatic contralateral limbs in patients undergoing revascularization for chronic limb-threatening ischemia (CLTI). METHODS We analyzed a database of 520 CLTI patients registered in a prospective, multicenter registry in Japan. Severe ischemia in asymptomatic contralateral limbs was determined as the Wound, Ischemia, and foot Infection (WIfI) classification system Ischemia (I) grade 2/3. RESULTS The prevalence of diabetes mellitus and dialysis-dependent renal failure was 74.2% and 53.5%, respectively. Asymptomatic limbs accounted for 65.0% [95% confidence interval (CI), 60.9-69.1%] of the overall population, and 55.0% (95% CI, 49.6-60.4%) of the asymptomatic contralateral limbs had WIfI I-2/3. The multivariate analysis identified age ≥65 years, dialysis-dependent renal failure, WIfI I-3 in the index limb, and loss of pressure sensation in the contralateral limb as independent risk factors for WIfI I-2/3 in asymptomatic contralateral limbs (all p < 0.05). The 3-year cumulative incidence rate of major adverse limb events (MALE) in asymptomatic contralateral limbs was 19.3% (95% CI, 15.1-23.7%), whereas that of all-cause mortality was 46.9% (95% CI, 41.0-52.5%). The corresponding rate including a composite of mortality and MALE was 58.8% (95% CI, 52.9-64.6%). In asymptomatic contralateral limbs, the adjusted hazard ratio of WIfI I-2/3 versus I-0/1 was 1.53 (95% CI, 1.11-2.10) for a composite of mortality and MALE, 1.96 (95% CI, 1.14-3.36) for MALE, and 1.37 (95% CI, 0.95-1.96) for mortality (p = 0.009, 0.015, and 0.091, respectively). CONCLUSIONS Two-thirds of CLTI patients had an asymptomatic contralateral limb, and approximately half of the asymptomatic contralateral limbs were exposed to severe ischemia. Older age, dialysis-dependent renal failure, WIfI I-3 in the index limb, and loss of pressure sensation in the contralateral limb were independently associated with severe ischemia in asymptomatic contralateral limbs. In addition to mortality, MALE commonly occurred in asymptomatic contralateral limbs, especially with WIfI I-2/3.
Collapse
|
25
|
Roy-Chowdhury E, Brauns N, Helmke A, Nordlohne J, Bräsen JH, Schmitz J, Volkmann J, Fleig SV, Kusche-Vihrog K, Haller H, von Vietinghoff S. Human CD16+ monocytes promote a pro-atherosclerotic endothelial cell phenotype via CX3CR1-CX3CL1 interaction. Cardiovasc Res 2021; 117:1510-1522. [PMID: 32717023 DOI: 10.1093/cvr/cvaa234] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/17/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022] Open
Abstract
AIMS Monocytes are central for atherosclerotic vascular inflammation. The human non-classical, patrolling subtype, which expresses high levels of CD16 and fractalkine receptor CX3CR1, strongly associates with cardiovascular events. This is most marked in renal failure, a condition with excess atherosclerosis morbidity. The underlying mechanism is not understood. This study investigated how human CD16+ monocytes modulate endothelial cell function. METHODS AND RESULTS In patients with kidney failure, CD16+ monocyte counts were elevated and dynamically decreased within a year after transplantation, chiefly due to a drop in CD14+CD16+ cells. The CX3CR1 ligand CX3CL1 was similarly elevated in the circulation of humans and mice with renal impairment. CX3CL1 up-regulation was also observed close to macrophage rich human coronary artery plaques. To investigate a mechanistic basis of this association, CD16+CX3CR1HIGH monocytes were co-incubated with primary human endothelium in vitro. Compared to classical CD14+ monocytes or transwell cocultures, CD16+ monocytes enhanced endothelial STAT1 and NF-κB p65 phosphorylation, up-regulated expression of CX3CL1 and interleukin-1β, numerous CCL and CXCL chemokines and molecules promoting leucocyte patrolling and adhesion such as ICAM1 and VCAM1. Genes required for vasodilatation including endothelial nitric oxide synthase decreased while endothelial collagen production increased. Uraemic patients' monocytes enhanced endothelial CX3CL1 even more markedly. Their receptor CX3CR1 was required for enhanced aortic endothelial stiffness in murine atherosclerosis with renal impairment. CX3CR1 dose-dependently modulated monocyte-contact-dependent gene expression in human endothelium. CONCLUSION By demonstrating endothelial proatherosclerotic gene regulation in direct contact with CD16+ monocytes, in part via cellular CX3CR1-CX3CL1 interaction, our data delineate a mechanism how this celltype can increase cardiovascular risk.
Collapse
Affiliation(s)
- Eva Roy-Chowdhury
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | - Nicolas Brauns
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | - Alexandra Helmke
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | - Johannes Nordlohne
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | | | - Jessica Schmitz
- Department of Pathology, Hannover Medical School, Hannover, Germany
| | - Julia Volkmann
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | - Susanne V Fleig
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | | | - Hermann Haller
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | - Sibylle von Vietinghoff
- Division of Nephrology and Hypertension, Department of Internal Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| |
Collapse
|
26
|
Hoogeveen RM, Verweij SL, Kaiser Y, Kroon J, Verberne HJ, Vogt L, Moens SJB, Stroes ESG. Atorvastatin treatment does not abolish inflammatory mediated cardiovascular risk in subjects with chronic kidney disease. Sci Rep 2021; 11:4126. [PMID: 33602971 PMCID: PMC7892998 DOI: 10.1038/s41598-021-83273-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/27/2021] [Indexed: 11/26/2022] Open
Abstract
Individuals with chronic kidney disease are at an increased risk for cardiovascular disease. This risk may partially be explained by a chronic inflammatory state in these patients, reflected by increased arterial wall and cellular inflammation. Statin treatment decreases cardiovascular risk and arterial inflammation in non-CKD subjects. In patients with declining kidney function, cardiovascular benefit resulting from statin therapy is attenuated, possibly due to persisting inflammation. In the current study, we assessed the effect of statin treatment on arterial wall and cellular inflammation. Fourteen patients with chronic kidney disease stage 3 or 4, defined by an estimated Glomerular Filtration Rate between 15 and 60 mL/min/1.73 m2, without cardiovascular disease were included in a single center, open label study to assess the effect of atorvastatin 40 mg once daily for 12 weeks (NTR6896). At baseline and at 12 weeks of treatment, we assessed arterial wall inflammation by 18F-fluoro-deoxyglucose positron-emission tomography computed tomography (18F-FDG PET/CT) and the phenotype of circulating monocytes were assessed. Treatment with atorvastatin resulted in a 46% reduction in LDL-cholesterol, but this was not accompanied by an attenuation in arterial wall inflammation in the aorta or carotid arteries, nor with changes in chemokine receptor expression of circulating monocytes. Statin treatment does not abolish arterial wall or cellular inflammation in subjects with mild to moderate chronic kidney disease. These results imply that CKD-associated inflammatory activity is mediated by factors beyond LDL-cholesterol and specific anti-inflammatory interventions might be necessary to further dampen the inflammatory driven CV risk in these subjects.
Collapse
Affiliation(s)
- Renate M Hoogeveen
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Simone L Verweij
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Hein J Verberne
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Liffert Vogt
- Department of Nephrology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Sophie J Bernelot Moens
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Rysz J, Franczyk B, Kujawski K, Sacewicz-Hofman I, Ciałkowska-Rysz A, Gluba-Brzózka A. Are Nutraceuticals Beneficial in Chronic Kidney Disease? Pharmaceutics 2021; 13:231. [PMID: 33562154 PMCID: PMC7915977 DOI: 10.3390/pharmaceutics13020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) is a worldwide health problem in which prevalence is constantly rising. The pathophysiology of CKD is complicated and has not been fully resolved. However, elevated oxidative stress is considered to play a vital role in the development of this disease. CKD is also thought to be an inflammatory disorder in which uremic toxins participate in the development of the inflammatory milieu. A healthy, balanced diet supports the maintenance of a good health status as it helps to reduce the risk of the development of chronic diseases, including chronic kidney disease, diabetes mellitus, and hypertension. Numerous studies have demonstrated that functional molecules and nutrients, including fatty acids and fiber as well as nutraceuticals such as curcumin, steviol glycosides, and resveratrol not only exert beneficial effects on pro-inflammatory and anti-inflammatory pathways but also on gut mucosa. Nutraceuticals have attracted great interest recently due to their potential favorable physiological effects on the human body and their safety. This review presents some nutraceuticals in which consumption could exert a beneficial impact on the development and progression of renal disease as well cardiovascular disease.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | - Krzysztof Kujawski
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| | | | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.); (K.K.)
| |
Collapse
|
28
|
Duraisamy P, Ravi S, Krishnan M, Livya CM, Manikandan B, Arunagirinathan K, Ramar M. Dynamic Role of Macrophage Sub Types for Development of Atherosclerosis and Potential Use of Herbal Immunomodulators as Imminent Therapeutic Strategy. Cardiovasc Hematol Agents Med Chem 2020; 20:2-12. [PMID: 33334298 DOI: 10.2174/1871525718666201217163207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis, a major contributor to cardiovascular disease is a global alarm causing mortality worldwide. Being a progressive disease in the arteries, it mainly causes recruitment of monocytes to the inflammatory sites and subside pathological conditions. Monocyte-derived macrophage mainly acts in foam cell formation by engorging the LDL molecules, oxidizes it into Ox-LDL and leads to plaque deposit development. Macrophages in general differentiate, proliferate and undergo apoptosis at the inflammatory site. Frequently two subtypes of macrophages M1 and M2 has to act crucially in balancing the micro-environmental conditions of endothelial cells in arteries. The productions of proinflammatory mediators like IL-1, IL-6, TNF-α by M1 macrophage has atherogenic properties majorly produced during the early progression of atherosclerotic plaques. To counteract cytokine productions and M1-M2 balance, secondary metabolites (phytochemicals) from plants act as a therapeutic agent in alleviating atherosclerosis progression. This review summarizes the fundamental role of the macrophage in atherosclerotic lesion formation along with its plasticity characteristic as well as recent therapeutic strategies using herbal components and anti-inflammatory cytokines as potential immunomodulators.
Collapse
Affiliation(s)
| | - Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai - 600 025. India
| | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai - 600 025. India
| | - Catherene M Livya
- Department of Zoology, University of Madras, Guindy Campus, Chennai - 600 025. India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai - 600 015. India
| | | | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai - 600 025. India
| |
Collapse
|
29
|
Oh CM, Park SK, Jung JY, Choi JM, Ha E, Lee EY, Kim JW, Kang HY, Yang HJ, Ryoo JH. Reduced Glomerular Filtration Rate and Risk of Stroke: A Nationwide Cohort Study in South Korea. J Atheroscler Thromb 2020; 28:928-941. [PMID: 33162421 PMCID: PMC8532060 DOI: 10.5551/jat.56143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aims:
Although chronic kidney disease is recognized as an independent risk factor for cerebrovascular disease, its association with hemorrhagic and ischemic stroke remains controversial.
Methods:
We conducted a retrospective cohort study using the National Health Insurance Service-National Sample Cohort, which is representative of the Korean population. A total of 195,772 Koreans who were not diagnosed with stroke before 2009 were included in this study from 2009 to 2013. The eGFR was divided into six categories (≥ 90, 75–89, 60–74, 45–59, 30–44, <30 mL/min/1.73 m
2
). The Kaplan–Meier plot was illustrated to compare the incidence of stroke. Cox proportional hazard model was used to estimate the hazard ratio (HR) of eGFR for risk of ischemic and hemorrhagic stroke by sex.
Results:
During an average of 4.36 years of follow-up period, 2,236 and 668 people were diagnosed with newly ischemic and hemorrhagic stroke, respectively. Age-adjusted incidence rate for ischemic stroke among people with eGFR <45 mL/min/1.73 m
2
was higher than those with eGFR ≥ 90 mL/min/1.73 m
2
, whereas that for hemorrhagic stroke among people with eGFR ≥ 90 mL/min/1.73 m
2
was higher than those with eGFR <45 mL/min/1.73 m
2
. After adjusting for multiple covariates, the adjusted HR for ischemic stroke increased with decreasing eGFR in men (
p
for trend <0.001), but not in women (
p
for trend=0.48). On the other hand, there was no significant relationship between eGFR and risk of hemorrhagic stroke in both men and women.
Conclusions:
Reduced glomerular filtration rate less than 45 mL/min/1.73 m
2
was associated with an increased risk of ischemic stroke, especially in men.
Collapse
Affiliation(s)
- Chang-Mo Oh
- Departments of Preventive Medicine, School of Medicine, Kyung Hee University
| | - Sung Keun Park
- Total healthcare center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of medicine
| | - Ju Young Jung
- Total healthcare center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of medicine
| | - Joong-Myung Choi
- Departments of Preventive Medicine, School of Medicine, Kyung Hee University
| | - Eunhee Ha
- Department of Occupational and Environment Medicine, College of Medicine, Ewha Womans University
| | - Eun-Young Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University
| | - Jung-Wook Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Kyung Hee University
| | - Hee Yong Kang
- Department of Anesthesiology and Pain Medicine, Kyung Hee University
| | - Hong Jun Yang
- Department of Preventive Medicine, Graduate School, Kyung Hee University
| | - Jae-Hong Ryoo
- Departments of Occupational and Environmental Medicine, School of Medicine, Kyung Hee University
| |
Collapse
|
30
|
Kim HY, Lee SJ, Hwang Y, Lee GH, Yoon CE, Kim HC, Yoo TH, Lee WW. Indoxyl Sulfate-Mediated Metabolic Alteration of Transcriptome Signatures in Monocytes of Patients with End-Stage Renal Disease (ESRD). Toxins (Basel) 2020; 12:toxins12100621. [PMID: 32998431 PMCID: PMC7601745 DOI: 10.3390/toxins12100621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 01/14/2023] Open
Abstract
End-stage renal disease (ESRD) is the final stage of chronic kidney disease, which is increasingly prevalent worldwide and is associated with the progression of cardiovascular disease (CVD). Indoxyl sulfate (IS), a major uremic toxin, plays a key role in the pathology of CVD via adverse effects in endothelial and immune cells. Thus, there is a need for a transcriptomic overview of IS responsive genes in immune cells of ESRD patients. Here, we investigated IS-mediated alterations in gene expression in monocytes from ESRD patients. Transcriptomic analysis of ESRD patient-derived monocytes and IS-stimulated monocytes from healthy controls was performed, followed by analysis of differentially expressed genes (DEGs) and gene ontology (GO). We found that 148 upregulated and 139 downregulated genes were shared between ESRD patient-derived and IS-stimulated monocytes. Interaction network analysis using STRING and ClueGo suggests that mainly metabolic pathways, such as the pentose phosphate pathway, are modified by IS in ESRD patient-derived monocytes. These findings were confirmed in IS-stimulated monocytes by the increased mRNA expression of genes including G6PD, PGD, and TALDO1. Our data suggest that IS causes alteration of metabolic pathways in monocytes of ESRD patients and, thus, these altered genes may be therapeutic targets.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea; (H.Y.K.); (Y.H.); (C.E.Y.)
- Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Su Jeong Lee
- Laboratory of Inflammation and Autoimmunity (LAI), Department of Biomedical Sciences and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea; (S.J.L.); (G.H.L.)
| | - Yuri Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea; (H.Y.K.); (Y.H.); (C.E.Y.)
| | - Ga Hye Lee
- Laboratory of Inflammation and Autoimmunity (LAI), Department of Biomedical Sciences and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea; (S.J.L.); (G.H.L.)
| | - Chae Eun Yoon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea; (H.Y.K.); (Y.H.); (C.E.Y.)
| | - Hyeon Chang Kim
- Cardiovascular and Metabolic Diseases Etiology Research Center and Department of Preventive Medicine, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Tae-Hyun Yoo
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea; (H.Y.K.); (Y.H.); (C.E.Y.)
- Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul 03080, Korea
- Laboratory of Inflammation and Autoimmunity (LAI), Department of Biomedical Sciences and BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea; (S.J.L.); (G.H.L.)
- Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul National University Hospital Biomedical Research Institute, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-2-740-8303; Fax: +82-2-743-0881
| |
Collapse
|
31
|
Podkowińska A, Formanowicz D. Chronic Kidney Disease as Oxidative Stress- and Inflammatory-Mediated Cardiovascular Disease. Antioxidants (Basel) 2020; 9:E752. [PMID: 32823917 PMCID: PMC7463588 DOI: 10.3390/antiox9080752] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Generating reactive oxygen species (ROS) is necessary for both physiology and pathology. An imbalance between endogenous oxidants and antioxidants causes oxidative stress, contributing to vascular dysfunction. The ROS-induced activation of transcription factors and proinflammatory genes increases inflammation. This phenomenon is of crucial importance in patients with chronic kidney disease (CKD), because atherosclerosis is one of the critical factors of their cardiovascular disease (CVD) and increased mortality. The effect of ROS disrupts the excretory function of each section of the nephron. It prevents the maintenance of intra-systemic homeostasis and leads to the accumulation of metabolic products. Renal regulatory mechanisms, such as tubular glomerular feedback, myogenic reflex in the supplying arteriole, and the renin-angiotensin-aldosterone system, are also affected. It makes it impossible for the kidney to compensate for water-electrolyte and acid-base disturbances, which progress further in the mechanism of positive feedback, leading to a further intensification of oxidative stress. As a result, the progression of CKD is observed, with a spectrum of complications such as malnutrition, calcium phosphate abnormalities, atherosclerosis, and anemia. This review aimed to show the role of oxidative stress and inflammation in renal impairment, with a particular emphasis on its influence on the most common disturbances that accompany CKD.
Collapse
Affiliation(s)
| | - Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland
| |
Collapse
|
32
|
Puvvula A, Jamthikar AD, Gupta D, Khanna NN, Porcu M, Saba L, Viskovic K, Ajuluchukwu JNA, Gupta A, Mavrogeni S, Turk M, Laird JR, Pareek G, Miner M, Sfikakis PP, Protogerou A, Kitas GD, Nicolaides A, Viswanathan V, Suri JS. Morphological Carotid Plaque Area Is Associated With Glomerular Filtration Rate: A Study of South Asian Indian Patients With Diabetes and Chronic Kidney Disease. Angiology 2020; 71:520-535. [PMID: 32180436 DOI: 10.1177/0003319720910660] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We evaluated the association between automatically measured carotid total plaque area (TPA) and the estimated glomerular filtration rate (eGFR), a biomarker of chronic kidney disease (CKD). Automated average carotid intima-media thickness (cIMTave) and TPA measurements in carotid ultrasound (CUS) were performed using AtheroEdge (AtheroPoint). Pearson correlation coefficient (CC) was then computed between the TPA and eGFR for (1) males versus females, (2) diabetic versus nondiabetic patients, and (3) between the left and right carotid artery. Overall, 339 South Asian Indian patients with either type 2 diabetes mellitus (T2DM) or CKD, or hypertension (stage 1 or stage 2) were retrospectively analyzed by acquiring cIMTave and TPA measurements of their left and right common carotid arteries (CCA; total CUS: 678, mean age: 54.2 ± 9.8 years; 75.2% males; 93.5% with T2DM). The CC between TPA and eGFR for different scenarios were (1) for males and females -0.25 (P < .001) and -0.35 (P < .001), respectively; (2) for T2DM and non-T2DM -0.26 (P < .001) and -0.49 (P = .02), respectively, and (3) for left and right CCA -0.25 (P < .001) and -0.23 (P < .001), respectively. Automated TPA is an equally reliable biomarker compared with cIMTave for patients with CKD (with or without T2DM) with subclinical atherosclerosis.
Collapse
Affiliation(s)
- Anudeep Puvvula
- Annu's Hospitals for Skin and Diabetes, Nellore, Andhra Pradesh, India
| | - Ankush D Jamthikar
- Department of Electronics and Communications Engineering, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra, India
| | - Deep Gupta
- Department of Electronics and Communications Engineering, Visvesvaraya National Institute of Technology, Nagpur, Maharashtra, India
| | - Narendra N Khanna
- Department of Cardiology, Indraprastha Apollo Hospitals, New Delhi, Delhi, India
| | - Michele Porcu
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), Cagliari, Italy
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, Zagreb, Croatia
| | | | - Ajay Gupta
- Department of Radiology, Weill Cornell Medicine, New York City, NY, USA
| | - Sophie Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Center, Athens, Greece
| | - Monika Turk
- Department of Neurology, University Medical Centre Maribor, Slovenia
| | - John R Laird
- Heart and Vascular Institute, Adventist Health St Helena, St Helena, CA, USA
| | - Gyan Pareek
- Minimally Invasive Urology Institute, Brown University, Providence, RI, USA
| | - Martin Miner
- Men's Health Center, Miriam Hospital, Providence, RI, USA
| | - Petros P Sfikakis
- Rheumatology Unit, National Kapodistrian University of Athens, Greece
| | - Athanasios Protogerou
- Department of Cardiovascular Prevention and Research Unit Clinic and Laboratory of Pathophysiology, National and Kapodistrian University of Athens, Greece
| | - George D Kitas
- R & D Academic Affairs, Dudley Group NHS Foundation Trust, Dudley, United Kingdom
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre and University of Nicosia Medical School, Cyprus
| | - Vijay Viswanathan
- M. V. Hospital for Diabetes and Professor M. Viswanathan Diabetes Research Centre, Chennai, Tamil Nadu, India
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint, Roseville, CA, USA
| |
Collapse
|
33
|
Engel JE, Chade AR. Macrophage polarization in chronic kidney disease: a balancing act between renal recovery and decline? Am J Physiol Renal Physiol 2019; 317:F1409-F1413. [PMID: 31566432 DOI: 10.1152/ajprenal.00380.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Macrophages are heterogenous cells of the innate immune system that can fluidly modulate their phenotype to respond to their local microenvironment. They are found throughout the renal compartments, where they contribute to homeostasis and function. However, renal injury activates molecular pathways that initially stimulate differentiation of macrophages into a proinflammatory M1 phenotype. Later in the course of healing, abundant apoptotic debris and anti-inflammatory cytokines induce the production of anti-inflammatory M2 macrophages, which contribute to tissue regeneration and repair. Thus, the dynamic balance of M1 and M2 populations may outline the burden of inflammation and process of tissue repair that define renal outcomes, which has been the impetus for therapeutic efforts targeting macrophages. This review will discuss the role of these phenotypes in the progression of chronic renal injury, potential pathogenic mechanisms, and the promise of macrophage-based therapeutic applications for chronic kidney disease.
Collapse
Affiliation(s)
- Jason E Engel
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alejandro R Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
34
|
Engel JE, Williams E, Williams ML, Bidwell GL, Chade AR. Targeted VEGF (Vascular Endothelial Growth Factor) Therapy Induces Long-Term Renal Recovery in Chronic Kidney Disease via Macrophage Polarization. Hypertension 2019; 74:1113-1123. [PMID: 31542966 DOI: 10.1161/hypertensionaha.119.13469] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) universally associates with renal microvascular rarefaction and inflammation, but whether a link exists between these 2 processes is unclear. We designed a therapeutic construct of VEGF (vascular endothelial growth factor) fused to an ELP (elastin-like polypeptide) carrier and show that it improves renal function in experimental renovascular disease. We test the hypothesis that ELP-VEGF therapy will improve CKD, and that recovery will be driven by decreasing microvascular rarefaction partly via modulation of macrophage phenotype and inflammation. CKD was induced in 14 pigs, which were observed for 14 weeks. At 6 weeks, renal blood flow and filtration were quantified using multidetector computed tomography, and then pigs received single intrarenal ELP-VEGF or placebo (n=7 each). Renal function was quantified again 4 and 8 weeks later. Pigs were euthanized and renal microvascular density, angiogenic and inflammatory markers, fibrosis, macrophage infiltration, and phenotype were quantified. Loss of renal hemodynamics in CKD was progressively recovered by ELP-VEGF therapy, accompanied by improved renal microvascular density, fibrosis, and expression of inflammatory mediators. Although renal macrophage infiltration was similar in both CKD groups, ELP-VEGF therapy distinctly shifted their phenotype from proinflammatory M1 to VEGF-expressing M2. Our study unravels potential mechanisms and feasibility of a new strategy to offset progression of CKD using drug-delivery technologies. The results indicate that renal recovery after ELP-VEGF therapy was largely driven by modulation of renal macrophages toward VEGF-expressing M2 phenotype, restoring VEGF signaling and sustaining improvement of renal function and microvascular integrity in CKD.
Collapse
Affiliation(s)
- Jason E Engel
- From the Departments of Physiology and Biophysics (J.E.E., E.W., M.L.W., A.R.C.), University of Mississippi Medical Center, Jackson
| | - Erika Williams
- From the Departments of Physiology and Biophysics (J.E.E., E.W., M.L.W., A.R.C.), University of Mississippi Medical Center, Jackson
| | - Maxx L Williams
- From the Departments of Physiology and Biophysics (J.E.E., E.W., M.L.W., A.R.C.), University of Mississippi Medical Center, Jackson
| | - Gene L Bidwell
- Neurology (G.L.B.), University of Mississippi Medical Center, Jackson.,Cell and Molecular Biology (G.L.B.), University of Mississippi Medical Center, Jackson.,Pharmacology and Toxicology (G.L.B.), University of Mississippi Medical Center, Jackson
| | - Alejandro R Chade
- From the Departments of Physiology and Biophysics (J.E.E., E.W., M.L.W., A.R.C.), University of Mississippi Medical Center, Jackson.,Medicine (A.R.C.), University of Mississippi Medical Center, Jackson.,Radiology (A.R.C.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
35
|
Schmit D, Fliser D, Speer T. Proprotein convertase subtilisin/kexin type 9 in kidney disease. Nephrol Dial Transplant 2019; 34:1266-1271. [DOI: 10.1093/ndt/gfz122] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
Chronic kidney disease (CKD) is associated with a substantially increased risk for the development of atherosclerotic cardiovascular (CV) disease. Accordingly, CV mortality is increased even in the earliest stages of CKD. In the general population and in CKD patients, high plasma levels of low-density lipoprotein cholesterol (LDL-C) are crucially involved in the initiation and progression of atherosclerotic vascular lesions. Lowering LDL-C by use of statins and/or ezetimibe represents the gold standard of lipid-lowering therapy, with a great body of evidence from several large clinical trials. Statin therapy reduces CV events in patients with normal and impaired kidney function alike, while the evidence for patients on maintenance haemodialysis is weaker. The inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) serine protease represents a novel lipid-lowering tool. Currently the monoclonal antibodies evolocumab and alirocumab are the approved PCSK9 inhibitors. Despite maximum-tolerated statin therapy, they efficiently further reduce LDL-C plasma levels without any major adverse effects. Moreover, in large clinical outcome trials, both antibodies have been proven to lower CV events. Notably, the LDL-lowering capacity was independent of baseline kidney function and also efficient in patients with moderate CKD. However, patients with severely impaired kidney function, that is, the population at the highest CV risk, have been excluded from those trials. The relevance of the LDL-independent effects of PCSK9 inhibitors, such as lowering lipoprotein(a) or ameliorating dyslipidaemia in patients with nephrotic syndrome, has to be determined. Therefore further specific studies assessing the effects and outcomes of PCSK9-inhibiting treatment in CKD patients are warranted.
Collapse
Affiliation(s)
- David Schmit
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Centre, Homburg/Saar, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Centre, Homburg/Saar, Germany
| | - Thimoteus Speer
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Centre, Homburg/Saar, Germany
| |
Collapse
|
36
|
Vlad C, Burlacu A, Florea L, Artene B, Badarau S, Covic A, Ureche C, Scripcariu D, Foia L, Covic A. A comprehensive review on apolipoproteins as nontraditional cardiovascular risk factors in end-stage renal disease: current evidence and perspectives. Int Urol Nephrol 2019; 51:1173-1189. [DOI: 10.1007/s11255-019-02170-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/09/2019] [Indexed: 12/17/2022]
|
37
|
Texakalidis P, Tzoumas A, Giannopoulos S, Jonnalagadda AK, Jabbour P, Rangel-Castilla L, Machinis T, Rivet DJ, Reavey-Cantwell J. Risk Factors for Restenosis After Carotid Revascularization: A Meta-Analysis of Hazard Ratios. World Neurosurg 2019; 125:414-424. [PMID: 30822589 DOI: 10.1016/j.wneu.2019.02.065] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 11/15/2022]
Abstract
BACKGROUND Carotid artery restenosis after carotid endarterectomy (CEA) or carotid artery stenting (CAS) will occur in 3%-30% of cases. Restenosis can lead to more frequent clinical and imaging monitoring and the potential for reoperation. We sought to define the demographic, clinical, and radiographic characteristics that influence the restenosis risk after carotid revascularization. METHODS The present study was performed in accordance with the PRISMA (preferred reporting items for systematic reviews and meta-analyses) guidelines. A random effects model meta-analysis of hazard ratios (HRs) was conducted. RESULTS Eighteen studies with 17,106 patients were included. Diabetes (HR, 1.68; 95% confidence interval [CI], 1.00-2.83; I2, 76.7%), dyslipidemia (HR, 1.77; 95% CI, 1.08-2.91; I2, 22.5%), female gender (HR, 1.50; 95% CI, 1.14-1.98, I2, 0%), chronic kidney disease (HR, 4.15; 95% CI, 1.69-10.19; I2, 44.5%), hypertension (HR, 1.99; 95% CI, 1.07-3.72; I2, 68%), smoking (HR, 1.65; 95% CI, 1.15-2.37; I2, 54.3%), and pretreatment stenosis >70% (HR, 1.04; 95% CI, 1.0-1.08; I2, 0%) showed a statistically significant increase in restenosis risk after carotid revascularization. Subgroup analyses of CEA and CAS showed that female gender and smoking status were significantly associated with recurrent stenosis after CEA but not after CAS. In contrast, hypertension was associated with restenosis after CAS but not after CEA. Patch endarterectomy (HR, 0.33; 95% CI, 0.22-0.50; I2, 0%) and symptomatic status at presentation in the CAS group (HR, 0.61; 95% CI, 0.41-0.90; I2, 0%) were associated with a decreased risk of restenosis. Antiplatelet use and coronary artery disease were not associated with restenosis risk. CONCLUSIONS Diabetes, dyslipidemia, female gender, renal failure, hypertension, and smoking were associated with an increased risk of restenosis, and patch endarterectomy and symptomatic status at presentation were associated with a decreased risk of carotid restenosis. Both female gender and current smoking status were only associated with recurrent stenosis after CEA, and hypertension was only associated with restenosis after CAS.
Collapse
Affiliation(s)
- Pavlos Texakalidis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA.
| | - Andreas Tzoumas
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Anil K Jonnalagadda
- Department of Cardiology, Medstar Washington Hospital Center, Washington District of Columbia, USA
| | - Pascal Jabbour
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Theofilos Machinis
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Dennis J Rivet
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia, USA
| | - John Reavey-Cantwell
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
38
|
Lazarova D, Shibata S, Ishii I, Zlateva G, Zhelev Z, Aoki I, Bakalova R. Imaging of redox-imbalance and oxidative stress in kidney in vivo, induced by dietary cholesterol. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1573153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Dessislava Lazarova
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Sayaka Shibata
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| | - Itsuko Ishii
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Chiba University Hospital, Chiba University, Chiba, Japan
| | - Genoveva Zlateva
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Zhivko Zhelev
- Department of Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| | - Rumiana Bakalova
- Department of Physics, Biophysics and Roentgenology, Medical Faculty, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
- Group of Quantum-State Controlled MRI, National Institute of Radiological Sciences (QST-NIRS), Chiba, Japan
| |
Collapse
|
39
|
Zeng L, Mathew AV, Byun J, Atkins KB, Brosius FC, Pennathur S. Myeloperoxidase-derived oxidants damage artery wall proteins in an animal model of chronic kidney disease-accelerated atherosclerosis. J Biol Chem 2018; 293:7238-7249. [PMID: 29581235 DOI: 10.1074/jbc.ra117.000559] [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] [Received: 10/26/2017] [Revised: 03/14/2018] [Indexed: 12/16/2022] Open
Abstract
Increased myeloperoxidase (MPO) levels and activity are associated with increased cardiovascular risk among individuals with chronic kidney disease (CKD). However, a lack of good animal models for examining the presence and catalytic activity of MPO in vascular lesions has impeded mechanistic studies into CKD-associated cardiovascular diseases. Here, we show for the first time that exaggerated atherosclerosis in a pathophysiologically relevant CKD mouse model is associated with increased macrophage-derived MPO activity. Male 7-week-old LDL receptor-deficient mice underwent sham (control mice) or 5/6 nephrectomy and were fed either a low-fat or high-fat, high-cholesterol diet for 24 weeks, and the extents of atherosclerosis and vascular reactivity were assessed. MPO expression and oxidation products-protein-bound oxidized tyrosine moieties 3-chlorotyrosine, 3-nitrotyrosine, and o,o'-dityrosine-were examined with immunoassays and confirmed with mass spectrometry (MS). As anticipated, the CKD mice had significantly higher plasma creatinine, urea nitrogen, and intact parathyroid hormone along with lower hematocrit and body weight. On both the diet regimens, CKD mice did not have hypertension but had lower cholesterol and triglyceride levels than the control mice. Despite the lower cholesterol levels, CKD mice had increased aortic plaque areas, fibrosis, and luminal narrowing. They also exhibited increased MPO expression and activity (i.e. increased oxidized tyrosines) that co-localized with infiltrating lesional macrophages and diminished vascular reactivity. In summary, unlike non-CKD mouse models of atherosclerosis, CKD mice exhibit increased MPO expression and catalytic activity in atherosclerotic lesions, which co-localize with lesional macrophages. These results implicate macrophage-derived MPO in CKD-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Lixia Zeng
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105
| | - Anna V Mathew
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105
| | - Jaeman Byun
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105
| | - Kevin B Atkins
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105
| | - Frank C Brosius
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48105
| | - Subramaniam Pennathur
- Department of Medicine, Division of Nephrology, Ann Arbor, Michigan 48105; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48105.
| |
Collapse
|
40
|
Ciccarelli G, D'Elia S, De Paulis M, Golino P, Cimmino G. Lipid Target in Very High-Risk Cardiovascular Patients: Lesson from PCSK9 Monoclonal Antibodies. Diseases 2018; 6:diseases6010022. [PMID: 29562587 PMCID: PMC5871968 DOI: 10.3390/diseases6010022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
The role of low-density lipoproteins (LDLs) as a major risk factor for cardiovascular disease has been demonstrated by several epidemiological studies. The molecular basis for LDLs in atherosclerotic plaque formation and progression is not completely unraveled yet. Pharmacological modulation of plasma LDL-C concentrations and randomized clinical trials addressing the impact of lipid-lowering interventions on cardiovascular outcome have clearly shown that reducing plasma LDL-C concentrations results in a significant decrease in major cardiovascular events. For many years, statins have represented the most powerful pharmacological agents available to lower plasma LDL-C concentrations. In clinical trials, it has been shown that the greater the reduction in plasma LDL-C concentrations, the lower the rate of major cardiovascular events, especially in high-risk patients, because of multiple risk factors and recurrent events. However, in a substantial number of patients, the recommended LDL target is difficult to achieve because of different factors: genetic background (familial hypercholesterolemia), side effects (statin intolerance), or high baseline plasma LDL-C concentrations. In the last decade, our understanding of the molecular mechanisms involved in LDL metabolism has progressed significantly and the key role of proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged. This protein is an enzyme able to bind the LDL receptors (LDL-R) on hepatocytes, favoring their degradation. Blocking PCSK9 represents an intriguing new therapeutic approach to decrease plasma LDL-C concentrations, which in recent studies has been demonstrated to also result in a significant reduction in major cardiovascular events.
Collapse
Affiliation(s)
- Giovanni Ciccarelli
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Saverio D'Elia
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Michele De Paulis
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| |
Collapse
|
41
|
Kaseda R, Tsuchida Y, Yang HC, Yancey PG, Zhong J, Tao H, Bian A, Fogo AB, Linton MRF, Fazio S, Ikizler TA, Kon V. Chronic kidney disease alters lipid trafficking and inflammatory responses in macrophages: effects of liver X receptor agonism. BMC Nephrol 2018; 19:17. [PMID: 29374468 PMCID: PMC5787279 DOI: 10.1186/s12882-018-0814-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/15/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Our aim was to evaluate lipid trafficking and inflammatory response of macrophages exposed to lipoproteins from subjects with moderate to severe chronic kidney disease (CKD), and to investigate the potential benefits of activating cellular cholesterol transporters via liver X receptor (LXR) agonism. METHODS LDL and HDL were isolated by sequential density gradient ultracentrifugation of plasma from patients with stage 3-4 CKD and individuals without kidney disease (HDLCKD and HDLCont, respectively). Uptake of LDL, cholesterol efflux to HDL, and cellular inflammatory responses were assessed in human THP-1 cells. HDL effects on inflammatory markers (MCP-1, TNF-α, IL-1β), Toll-like receptors-2 (TLR-2) and - 4 (TLR-4), ATP-binding cassette class A transporter (ABCA1), NF-κB, extracellular signal regulated protein kinases 1/2 (ERK1/2) were assessed by RT-PCR and western blot before and after in vitro treatment with an LXR agonist. RESULTS There was no difference in macrophage uptake of LDL isolated from CKD versus controls. By contrast, HDCKD was significantly less effective than HDLCont in accepting cholesterol from cholesterol-enriched macrophages (median 20.8% [IQR 16.1-23.7] vs control (26.5% [IQR 19.6-28.5]; p = 0.008). LXR agonist upregulated ABCA1 expression and increased cholesterol efflux to HDL of both normal and CKD subjects, although the latter continued to show lower efflux capacity. HDLCKD increased macrophage cytokine response (TNF-α, MCP-1, IL-1β, and NF-κB) versus HDLCont. The heightened cytokine response to HDLCKD was further amplified in cells treated with LXR agonist. The LXR-augmentation of inflammation was associated with increased TLR-2 and TLR-4 and ERK1/2. CONCLUSIONS Moderate to severe impairment in kidney function promotes foam cell formation that reflects impairment in cholesterol acceptor function of HDLCKD. Activation of cellular cholesterol transporters by LXR agonism improves but does not normalize efflux to HDLCKD. However, LXR agonism actually increases the pro-inflammatory effects of HDLCKD through activation of TLRs and ERK1/2 pathways.
Collapse
Affiliation(s)
- Ryohei Kaseda
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Yohei Tsuchida
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Hai-Chun Yang
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | | | - Jianyong Zhong
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Huan Tao
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Aihua Bian
- Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Agnes B. Fogo
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Mac Rae F. Linton
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Oregon, Portland USA
| | | | - Valentina Kon
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| |
Collapse
|
42
|
Nordlohne J, Helmke A, Ge S, Rong S, Chen R, Waisman A, Haller H, von Vietinghoff S. Aggravated Atherosclerosis and Vascular Inflammation With Reduced Kidney Function Depend on Interleukin-17 Receptor A and Are Normalized by Inhibition of Interleukin-17A. JACC Basic Transl Sci 2018; 3:54-66. [PMID: 30062194 PMCID: PMC6058956 DOI: 10.1016/j.jacbts.2017.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/25/2022]
Abstract
Moderate renal impairment significantly increases atherosclerotic lesion size and leukocyte numbers, most markedly macrophages and T cells, in LDLr–/– mice. IL-17 receptor A–deficient LDLr–/– mice are protected from the growth in lesion size and leukocyte infiltrate in renal impairment. Monocytes, especially Ly6C/GR1HIGH cells, express high levels of IL-17 receptor A. IL-17A increases monocyte adhesion to the aortic wall and enhances endothelial cell pro-inflammatory cytokine production. Ablation of IL-17A or IL-17A blockade normalizes the inflammatory aortic wall infiltrate even in established atherosclerosis.
Effective therapy of atherosclerotic complications in patients with chronic kidney disease (CKD) is an unmet clinical need. Cardiovascular events are the most common cause of death. At a glomerular filtration rate ≤60 ml/min, these events are increased also after correction for common risk factors. Previous studies have reported enhanced vascular inflammation in mice and recently also in humans. Our current data show, in a mouse model of atherosclerosis in moderate renal impairment, that interleukin-17 receptor A is instrumental in this condition, and blockade of this pathway can normalize arterial inflammation even in advanced atherosclerosis.
Collapse
Affiliation(s)
- Johannes Nordlohne
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Alexandra Helmke
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Shuwang Ge
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Department of Nephrology, Tongji Hospital, Huazhong University of Science and Technology, Huazhong, People's Republic of China
| | - Song Rong
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Rongjun Chen
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University of Mainz, Mainz, Germany
| | - Hermann Haller
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Sibylle von Vietinghoff
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
43
|
Keating ST, van Diepen JA, Riksen NP, El-Osta A. Epigenetics in diabetic nephropathy, immunity and metabolism. Diabetologia 2018; 61:6-20. [PMID: 29128937 PMCID: PMC6448927 DOI: 10.1007/s00125-017-4490-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/22/2017] [Indexed: 01/01/2023]
Abstract
When it comes to the epigenome, there is a fine line between clarity and confusion-walk that line and you will discover another fascinating level of transcription control. With the genetic code representing the cornerstone of rules for information that is encoded to proteins somewhere above the genome level there is a set of rules by which chemical information is also read. These epigenetic modifications show a different side of the genetic code that is diverse and regulated, hence modifying genetic transcription transiently, ranging from short- to long-term alterations. While this complexity brings exquisite control it also poses a formidable challenge to efforts to decode mechanisms underlying complex disease. Recent technological and computational advances have improved unbiased acquisition of epigenomic patterns to improve our understanding of the complex chromatin landscape. Key to resolving distinct chromatin signatures of diabetic complications is the identification of the true physiological targets of regulatory proteins, such as reader proteins that recognise, writer proteins that deposit and eraser proteins that remove specific chemical moieties. But how might a diverse group of proteins regulate the diabetic landscape from an epigenomic perspective? Drawing from an ever-expanding compendium of experimental and clinical studies, this review details the current state-of-play and provides a perspective of chromatin-dependent mechanisms implicated in diabetic complications, with a special focus on diabetic nephropathy. We hypothesise a codified signature of the diabetic epigenome and provide examples of prime candidates for chemical modification. As for the pharmacological control of epigenetic marks, we explore future strategies to expedite and refine the search for clinically relevant discoveries. We also consider the challenges associated with therapeutic strategies targeting epigenetic pathways.
Collapse
Affiliation(s)
- Samuel T Keating
- Department of Internal Medicine, Department of Internal Medicine (463), Radboud University Medical Center, Nijmegen, PO Box 9101, 6500 HB, Nijmegen, the Netherlands.
| | - Janna A van Diepen
- Department of Internal Medicine, Department of Internal Medicine (463), Radboud University Medical Center, Nijmegen, PO Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Department of Internal Medicine (463), Radboud University Medical Center, Nijmegen, PO Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Assam El-Osta
- Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Pathology, The University of Melbourne, Parkville, VIC, Australia.
- Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
44
|
Kakuta K, Dohi K, Miyoshi M, Yamanaka T, Kawamura M, Masuda J, Kurita T, Ogura T, Yamada N, Sumida Y, Ito M. Impact of renal function on the underlying pathophysiology of coronary plaque composition in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2017; 16:131. [PMID: 29025416 PMCID: PMC5639771 DOI: 10.1186/s12933-017-0618-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/06/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Both the progression of diabetic kidney disease and increased glycemic variability play important roles in the pathogenesis of coronary plaque formation via inflammatory pathways in patients with type 2 diabetes mellitus (T2DM). Therefore we evaluated the role of renal function in the contributory effects of blood glucose fluctuations and blood levels of inflammatory cytokine concentrations on the tissue characteristics of coronary plaques in patients with T2DM. METHODS We prospectively enrolled 71 T2DM patients (mean age: 68 ± 9, male 79%) with 153 coronary artery lesions. Patients were divided into 2 groups according to their estimated glomerular filtration rate (eGFR) levels: Group 1 (≥ 60 mL/min/1.73 m2, n = 40) and Group 2 (< 60 mL/min/1.73 m2, n = 31). All patients underwent continuous glucose monitoring (CGM) for 120 h and the mean amplitude of glycemic excursions (MAGE) was calculated. Serum tumor necrosis factor (TNF)-α was also measured. In addition, gray-scale coronary intravascular ultrasound (IVUS) and iMap-IVUS were performed in the coronary lesions with < 50% luminal reduction. RESULTS In Group 1, MAGE correlated with percent lipidic volume (%LV) (r = 0.477, p = 0.002). In this group, stepwise multivariate linear regression analyses showed that only MAGE was independently associated with %LV (β = 0.477, p = 0.002). In contrast, in Group 2, only serum TNF-α correlated with percent fibrotic volume (%FV) (r = - 0.471, p = 0.007), %LV (r = 0.496, p = 0.005) and percent necrotic volume (%NV) (r = 0.426, p = 0.017). In this group, stepwise multivariate linear regression analyses showed that only serum TNF-α was independently associated with each tissue characteristic (%FV β = - 0.471 and p = 0.007, %LV β = 0.496 and p = 0.005, %NV: β = 0.426 and p = 0.017). CONCLUSIONS In T2DM patients, the tissue characteristics of coronary plaques were associated with MAGE in patients with eGFR ≥ 60 mL/min/1.73 m2 and with serum TNF-α in those with eGFR < 60 mL/min/1.73 m2.
Collapse
Affiliation(s)
- Kentaro Kakuta
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan.,Department of Cardiology, JCHO Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Kaoru Dohi
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan.
| | - Miho Miyoshi
- Department of Diabetes and Endocrinology, JCHO Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Takashi Yamanaka
- Department of Cardiology, JCHO Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Masaki Kawamura
- Department of Cardiology, JCHO Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Jun Masuda
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Tairo Kurita
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Toru Ogura
- Clinical Research Support Center, Mie University Hospital, Tsu, Japan
| | - Norikazu Yamada
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Yasuhiro Sumida
- Department of Diabetes and Endocrinology, JCHO Yokkaichi Hazu Medical Center, Yokkaichi, Japan
| | - Masaaki Ito
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| |
Collapse
|
45
|
Shen J, Rasmussen M, Dong QR, Tepel M, Scholze A. Expression of the NRF2 Target Gene NQO1 Is Enhanced in Mononuclear Cells in Human Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9091879. [PMID: 28785379 PMCID: PMC5530440 DOI: 10.1155/2017/9091879] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Reduced nuclear factor erythroid 2-related factor 2 (NRF2) pathway activity was reported in models of chronic kidney disease (CKD). Pharmacological activation of NRF2 is supposed to improve renal function, but data concerning the NRF2 activity in human CKD are lacking. We investigated the NRF2 target NAD(P)H:quinone oxidoreductase 1 (NQO1) as a readout parameter for NRF2 activity in monocytes of CKD patients (n = 63) compared to those of healthy controls (n = 16). The NQO1 gene expression was quantified using real-time PCR and the protein content by in-cell Western assays. We found a 3-4-fold increase in NQO1 gene expression in CKD 1-5 (n = 29; 3.5 for NQO1/ribosomal protein L41; p < 0.001). This was accompanied by a 1.1-fold increase in NQO1 protein (p = 0.06). Cardiovascular disease prevalence was higher in CKD 1-5 patients with higher compared to those with lower NQO1 gene expression (p = 0.02). In advanced uremia, in dialysis patients (n = 34), NQO1 gene expression was less robustly upregulated than that in CKD 1-5, while NQO1 protein was not upregulated. We conclude that in mononuclear cells of CKD patients, the NRF2 pathway is activated by coexisting pathogenic mechanisms, but in advanced uremia, the effectiveness of this upregulation is reduced. Both processes could interfere with pharmacological NRF2 activation.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | | | - Qi-Rong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Martin Tepel
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Alexandra Scholze
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
Rubio-Navarro A, Guerrero-Hue M, Martín-Fernandez B, Cortegano I, Olivares-Alvaro E, de Las Heras N, Alía M, de Andrés B, Gaspar ML, Egido J, Moreno JA. Phenotypic Characterization of Macrophages from Rat Kidney by Flow Cytometry. J Vis Exp 2016. [PMID: 27805599 DOI: 10.3791/54599] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is increasing evidence suggesting the important role of inflammation and, subsequently, macrophages in the development and progression of renal disease. Macrophages are heterogeneous cells that have been implicated in kidney injury. Macrophages may be classified into two different phenotypes: classically activated macrophages (M1 macrophages), that release pro-inflammatory cytokines and promote fibrosis; and alternatively activated macrophages (M2 macrophages) that are associated with immunoregulatory and tissue-remodeling functions. These macrophage phenotypes need to be discriminated and analyzed to determine their contribution to renal injury. However, there are scarce studies reporting consistent phenotypic and functional information about macrophage subtypes in inflammatory renal disease models, especially in rats. This fact may be related to the limited macrophage markers used in rats, contrary to mice. Therefore, novel strategies are necessary to quantify and characterize the renal content of these infiltrating cells in a reliable way. This manuscript details a protocol for kidney digestion and further phenotypic and quantitative analysis of macrophages from rat kidneys by flow cytometry. Briefly, kidneys were incubated with collagenase and total macrophages were identified according to the dual presence of CD45 (leukocytes common antigen) and CD68 (PAN macrophage marker) in live cells.This was followed by surface staining of CD86 (M1 marker) and CD163 (M2 marker). Rat peritoneal macrophages were used as positive control for macrophage marker detection by flow cytometry. Our protocol resulted in low cellular mortality and allowed characterization of different intracellular and surface protein markers, thus limiting the loss of cellular integrity observed in other protocols. Moreover, this procedure allows the use of macrophages for further techniques, including cell sorting and mRNA or protein expression studies, among others.
Collapse
Affiliation(s)
- Alfonso Rubio-Navarro
- Renal, Vascular and Diabetes Research Lab, IIS-Fundaciòn Jiménez Dìaz, Autonoma University
| | - Melania Guerrero-Hue
- Renal, Vascular and Diabetes Research Lab, IIS-Fundaciòn Jiménez Dìaz, Autonoma University
| | | | - Isabel Cortegano
- Department of Immunology, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII)
| | | | | | - Mario Alía
- Department of Immunology, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII)
| | - Belén de Andrés
- Department of Immunology, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII)
| | - María Luisa Gaspar
- Department of Immunology, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII)
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Lab, IIS-Fundaciòn Jiménez Dìaz, Autonoma University
| | - Juan Antonio Moreno
- Renal, Vascular and Diabetes Research Lab, IIS-Fundaciòn Jiménez Dìaz, Autonoma University;
| |
Collapse
|
47
|
Lower Superoxide Dismutase 2 (SOD2) Protein Content in Mononuclear Cells Is Associated with Better Survival in Patients with Hemodialysis Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7423249. [PMID: 27630759 PMCID: PMC5007362 DOI: 10.1155/2016/7423249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 11/18/2022]
Abstract
Mitochondrial superoxide dismutase 2 (SOD2) converts superoxide anions to hydrogen peroxide and oxygen. Human data on SOD2 protein content in chronic kidney disease (CKD) are sparse and mortality data are lacking. We investigated SOD2 protein content in monocytes from patients with hemodialysis therapy (n = 81), CKD stage 1-5 (n = 120), and healthy controls (n = 13) using in-cell Western assays. SOD2 protein decreased from CKD stage 1 until stage 4 whereas it increased again in stage 5 with and without hemodialysis. SOD2 gene expression, analyzed by quantitative real-time PCR, was not significantly different between the groups. Elevating cellular superoxide production reduced SOD2 protein content. This effect was abolished by the superoxide dismutase mimetic Tempol. Using gelelectrophoresis and Western blot we did not detect nitrotyrosine modifications of SOD2 in CKD. Finally, in patients with CKD stage 5 with hemodialysis therapy higher than median SOD2 protein content was associated with higher all-cause mortality. In conclusion, SOD2 protein content declined in CKD until stage 4 while SOD2 gene expression did not. Increased cellular superoxide anion production might affect SOD2 protein content. In advanced CKD (stage 5) SOD2 protein content increased again, but higher than median SOD2 protein content in these patients did not confer a survival benefit.
Collapse
|
48
|
Dong L, Nordlohne J, Ge S, Hertel B, Melk A, Rong S, Haller H, von Vietinghoff S. T Cell CX3CR1 Mediates Excess Atherosclerotic Inflammation in Renal Impairment. J Am Soc Nephrol 2016; 27:1753-64. [PMID: 26449606 PMCID: PMC4884117 DOI: 10.1681/asn.2015050540] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 08/24/2015] [Indexed: 12/27/2022] Open
Abstract
Reduced kidney function increases the risk for atherosclerosis and cardiovascular death. Leukocytes in the arterial wall contribute to atherosclerotic plaque formation. We investigated the role of fractalkine receptor CX3CR1 in atherosclerotic inflammation in renal impairment. Apoe(-/-) (apolipoprotein E) CX3CR1(-/-) mice with renal impairment were protected from increased aortic atherosclerotic lesion size and macrophage accumulation. Deficiency of CX3CR1 in bone marrow, only, attenuated atherosclerosis in renal impairment in an independent atherosclerosis model of LDL receptor-deficient (LDLr(-/-)) mice as well. Analysis of inflammatory leukocytes in atherosclerotic mixed bone-marrow chimeric mice (50% wild-type/50% CX3CR1(-/-) bone marrow into LDLr(-/-) mice) showed that CX3CR1 cell intrinsically promoted aortic T cell accumulation much more than CD11b(+)CD11c(+) myeloid cell accumulation and increased IL-17-producing T cell counts. In vitro, fewer TH17 cells were obtained from CX3CR1(-/-) splenocytes than from wild-type splenocytes after polarization with IL-6, IL-23, and TGFβ Polarization of TH17 or TREG cells, or stimulation of splenocytes with TGFβ alone, increased T cell CX3CR1 reporter gene expression. Furthermore, TGFβ induced CX3CR1 mRNA expression in wild-type cells in a dose- and time-dependent manner. In atherosclerotic LDLr(-/-) mice, CX3CR1(+/-) T cells upregulated CX3CR1 and IL-17A production in renal impairment, whereas CX3CR1(-/-) T cells did not. Transfer of CX3CR1(+/-) but not Il17a(-/-) T cells into LDLr(-/-)CX3CR1(-/-) mice increased aortic lesion size and aortic CD11b(+)CD11c(+) myeloid cell accumulation in renal impairment. In summary, T cell CX3CR1 expression can be induced by TGFβ and is instrumental in enhanced atherosclerosis in renal impairment.
Collapse
Affiliation(s)
- Lei Dong
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany; Department of Nephrology, Tongji Hospital, Huazhong University of Science and Technology, China; and
| | - Johannes Nordlohne
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Shuwang Ge
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany; Department of Nephrology, Tongji Hospital, Huazhong University of Science and Technology, China; and
| | - Barbara Hertel
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Anette Melk
- Division of Pediatrics, Hannover Medical School, Hannover, Germany
| | - Song Rong
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
49
|
Helmke A, von Vietinghoff S. Extracellular vesicles as mediators of vascular inflammation in kidney disease. World J Nephrol 2016; 5:125-38. [PMID: 26981436 PMCID: PMC4777783 DOI: 10.5527/wjn.v5.i2.125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/18/2015] [Accepted: 01/08/2016] [Indexed: 02/06/2023] Open
Abstract
Vascular inflammation is a common cause of renal impairment and a major cause of morbidity and mortality of patients with kidney disease. Current studies consistently show an increase of extracellular vesicles (EVs) in acute vasculitis and in patients with atherosclerosis. Recent research has elucidated mechanisms that mediate vascular wall leukocyte accumulation and differentiation. This review addresses the role of EVs in this process. Part one of this review addresses functional roles of EVs in renal vasculitis. Most published data address anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis and indicate that the number of EVs, mostly of platelet origin, is increased in active disease. EVs generated from neutrophils by activation by ANCA can contribute to vessel damage. While EVs are also elevated in other types of autoimmune vasculitis with renal involvement such as systemic lupus erythematodes, functional consequences beyond intravascular thrombosis remain to be established. In typical hemolytic uremic syndrome secondary to infection with shiga toxin producing Escherichia coli, EV numbers are elevated and contribute to toxin distribution into the vascular wall. Part two addresses mechanisms how EVs modulate vascular inflammation in atherosclerosis, a process that is aggravated in uremia. Elevated numbers of circulating endothelial EVs were associated with atherosclerotic complications in a number of studies in patients with and without kidney disease. Uremic endothelial EVs are defective in induction of vascular relaxation. Neutrophil adhesion and transmigration and intravascular thrombus formation are critically modulated by EVs, a process that is amenable to therapeutic interventions. EVs can enhance monocyte adhesion to the endothelium and modulate macrophage differentiation and cytokine production with major influence on the local inflammatory milieu in the plaque. They significantly influence lipid phagocytosis and antigen presentation by mononuclear phagocytes. Finally, platelet, erythrocyte and monocyte EVs cooperate in shaping adaptive T cell immunity. Future research is needed to define changes in uremic EVs and their differential effects on inflammatory leukocytes in the vessel wall.
Collapse
|
50
|
Pitavastatin Reduces Inflammation in Atherosclerotic Plaques in Apolipoprotein E-Deficient Mice with Late Stage Renal Disease. PLoS One 2015; 10:e0138047. [PMID: 26367531 PMCID: PMC4569429 DOI: 10.1371/journal.pone.0138047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 08/24/2015] [Indexed: 12/20/2022] Open
Abstract
Objectives Chronic renal disease (CRD) accelerates atherosclerosis and cardiovascular calcification. Statins reduce low-density lipoprotein-cholesterol levels in patients with CRD, however, the benefits of statins on cardiovascular disease in CRD remain unclear. This study has determined the effects of pitavastatin, the newest statin, on arterial inflammation and calcification in atherogenic mice with CRD. Methods and Results CRD was induced by 5/6 nephrectomy in cholesterol-fed apolipoprotein E-deficient mice. Mice were randomized into three groups: control mice, CRD mice, and CRD mice treated with pitavastatin. Ultrasonography showed that pitavastatin treatment significantly attenuated luminal stenosis in brachiocephalic arteries of CRD mice. Near-infrared molecular imaging and correlative Mac3 immunostaining demonstrated a significant reduction in macrophage accumulation in pitavastatin-treated CRD mice. Pitavastatin treatment reduced levels of osteopontin in plasma and atherosclerotic lesions in CRD mice, but did not produce a significant reduction in calcification in atherosclerotic plaques as assesses by histology. CRD mice had significantly higher levels of phosphate in plasma than did control mice, which did not change by pitavastatin. In vitro, pitavastatin suppressed the expression of osteopontin in peritoneal macrophages stimulated with phosphate or calcium/phosphate in concentrations similar to those found in human patients with CRD. Conclusion Our study provides in vivo evidence that pitavastatin reduces inflammation within atherosclerotic lesions in CRD mice.
Collapse
|