1
|
Liu H. Association between PCSK9 inhibitors and acute kidney injury: a pharmacovigilance study. Front Pharmacol 2024; 15:1353848. [PMID: 39148544 PMCID: PMC11324468 DOI: 10.3389/fphar.2024.1353848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/27/2024] [Indexed: 08/17/2024] Open
Abstract
Background PCSK9 inhibitors are a novel class of lipid-lowering medications, and numerous clinical studies have confirmed their significant role in improving the progression of chronic kidney disease. However, recent case reports have indicated new evidence regarding their association with acute kidney injury (AKI), with some patients experiencing acute tubular injury after PCSK9 inhibitors use. Objectives To clarify the relationship between PCSK9 inhibitors and AKI, we conducted a pharmacovigilance study. Methods Using the Food and Drug Administration Adverse Event Reporting System (FAERS) database from the third quarter of 2015 to the fourth quarter of 2022, a disproportionality analysis was employed to identify adverse events suggestive of AKI after PCSK9 inhibitors use. The drugs of interest included evolocumab and alirocumab. Results A total of 144,341 adverse event reports related to PCSK9 inhibitors were analyzed, among which 444 cases were suspected of AKI for evolocumab, and 172 cases for alirocumab. Evolocumab had a greater impact on AKI in males (ROR 1.4, 95% CI 1.54-1.69). The ROR and 95% CI for evolocumab and Alirocumab were 0.13 (0.12-0.14) and 0.26 (0.23-0.30) respectively. Further analysis of AKI associated with the concomitant use of PCSK9 inhibitors with cephalosporins, furosemide, torsemide, pantoprazole, omeprazole, and esomeprazole revealed ROR and 95% CI of 0.38 (0.23-0.62), 0.38 (0.31-0.48), 0.18 (0.08-0.38), 0.23 (0.17-0.29), 0.20 (0.16-0.26), and 0.14 (0.10-0.20) respectively. Conclusion Through the FAERS database, we analyzed the clinical characteristics of AKI associated with PCSK9 inhibitors, exploring its risks. Our findings suggest that PCSK9 inhibitors might have a potential protective effect against AKI and exhibit similar effects when co-administered with other nephrotoxic drugs.
Collapse
Affiliation(s)
- Hailing Liu
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
2
|
Ji X, Guo HY, Han M, Peng H, Yuan H. Association between genetically proxied PCSK9 inhibition and systemic lupus erythematosus risk: A mendelian randomization study. Int J Rheum Dis 2024; 27:e15106. [PMID: 38568054 DOI: 10.1111/1756-185x.15106] [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: 10/18/2023] [Revised: 12/13/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Preclinical and epidemiological studies suggest that proprotein convertase subtilisin/kexin type 9 (PCSK9) had a potential effect on the development of SLE, but it was unclear whether a causal relationship exists. We aimed to investigate the association between genetically proxied inhibition of PCSK9 and the risk of SLE using a two-sample Mendelian randomization (MR) approach. METHODS Single nucleotide polymorphisms (SNPs) associated with PCSK9 were extracted from pooled data obtained from the Global Lipid Genetics Consortium (GLGC) Genome-wide Association Study (GWAS) related to LDL-c levels, which was used as a proxy for PCSK9 inhibition. Pooled statistics for SLE were obtained from an independent GWAS dataset including 5201 SLE patients and 9066 controls. Inverse variance-weighted random-effects models were used to examine the association between genetically proxied inhibition of PCSK9 and the risk of SLE. MR-Egger, weighted median, weighted mode, Simple mode, and co-location analyses were used as sensitivity analyses to test the robustness of the analyses. RESULTS Genetically proxied inhibition of PCSK9 was associated with a reduced risk of SLE (OR = 0.51, 95% CI = 0.34 to 0.77, p = .001). This finding was replicated in an earlier GLGC GWAS analysis (OR = 0.59, 95% CI = 0.40 to 0.87, p = .007). Sensitivity analysis ensured that the results were robust. Co-localization analysis did not find evidence of shared causal variation between PCSK9 and SLE. CONCLUSIONS This Mendelian randomization study showed that PCSK9 was associated with SLE pathogenesis, and its inhibition was associated with a reduced risk of SLE. This study has offered a prospective therapeutic avenue for intervening in the progression of SLE by inhibiting PCSK9 levels.
Collapse
Affiliation(s)
- Xincan Ji
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Hao-Yang Guo
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Mengqi Han
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Hui Peng
- Science and Technology Department, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hui Yuan
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
3
|
Li J, Li J, Lin C, Zhou J, Wang J, Wang F, Li H, Zhou Z. Genetically proxied PCSK9 inhibition is associated with reduced psoriatic arthritis risk. Inflamm Res 2024; 73:475-484. [PMID: 38341813 PMCID: PMC10894168 DOI: 10.1007/s00011-024-01850-3] [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: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Lipid pathways play a crucial role in psoriatic arthritis development, and some lipid-lowering drugs are believed to have therapeutic benefits due to their anti-inflammatory properties. Traditional observational studies face issues with confounding factors, complicating the interpretation of causality. This study seeks to determine the genetic link between these medications and the risk of psoriatic arthritis. METHODS This drug target study utilized the Mendelian randomization strategy. We harnessed high-quality data from population-level genome-wide association studies sourced from the UK Biobank and FinnGen databases. The inverse variance-weighted method, complemented by robust pleiotropy methods, was employed. We examined the causal relationships between three lipid-lowering agents and psoriatic arthritis to unveil the underlying mechanisms. RESULTS A significant association was observed between genetically represented proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition and a decreased risk of psoriatic arthritis (odds ratio [OR]: 0.51; 95% CI 0.14-0.88; P < 0.01). This association was further corroborated in an independent dataset (OR 0.60; 95% CI 0.25-0.94; P = 0.03). Sensitivity analyses affirmed the absence of statistical evidence for pleiotropic or genetic confounding biases. However, no substantial associations were identified for either 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors or Niemann-Pick C1-like 1 inhibitors. CONCLUSIONS This Mendelian randomization analysis underscores the pivotal role of PCSK9 in the etiology of psoriatic arthritis. Inhibition of PCSK9 is associated with reduced psoriatic arthritis risk, highlighting the potential therapeutic benefits of existing PCSK9 inhibitors.
Collapse
Affiliation(s)
- Junhong Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
- Department of Orthopaedics and Trauma, The Affiliated Hospital of Yunnan University, Yunnan University, Kunming, 650091, China
| | - Jianfeng Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
| | - Chengkai Lin
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
| | - Jiaxiang Zhou
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jianmin Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
- Department of Spinal Surgery, Yantaishan Hospital, Yantai, 264003, China
| | - Fuan Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
| | - Haizhen Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Guangming District, The Seventh Affiliated Hospital, Sun Yat-Sen University, 66 Gongchang Road, Shenzhen, 518107, China.
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Zhu H, Meng Q, Liu X, Zhai C, Sun J, Wang R, Xu L, Yang X. Association of circulating proprotein convertase subtilisin/kexin type 9 concentration with coagulation abnormalities in patients with primary membranous nephropathy. Ren Fail 2023; 45:2212084. [PMID: 37183799 DOI: 10.1080/0886022x.2023.2212084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVES The aims of the study were to explore the potential associations between plasma proprotein convertase subtilisin/kexin type 9 (PCSK9) and coagulation indexes in patients with primary membranous nephropathy (PMN). METHODS A total of 87 patients diagnosed with PMN were enrolled in our study. 30 healthy participants were recruited to match PMN participants. Plasma PCSK9 concentrations were tested by enzyme-linked immunosorbent assay (ELISA). Correlations between PCSK9 and coagulation abnormalities in patients with PMN were analyzed using univariate and multiple linear regression analysis. RESULTS Plasma PCSK9 levels in patients with PMN were significantly higher than that in healthy controls [232.0 (143.5, 359.5) ng/mL vs. 166.8 (129.7, 199.7) ng/mL; p = 0.001]. Plasma levels of PCSK9 were positively correlated with factor VIII, factor IX, factor XI, log-transformed tissue factor, protein C and protein S (r = 0.267, p = 0.013; r = 0.496, p < 0.001; r = 0.217, p = 0.045; r = 0.584, p < 0.001; r = 0.372, p = 0.001; r = 0.282, p = 0.011). In multiple linear regression analysis, PCSK9 concentration was independently and positively correlated with factor VIII, factor IX, and tissue factor (β = 0.186, p = 0.047; β = 0.325, p = 0.001; β = 0.531, p < 0.001; respectively). PCSK9 concentration was independently and negatively correlated with PT (β= -0.343, p = 0.011). CONCLUSION Plasma PCSK9 levels had good positive correlations with procoagulant clotting factors and negative correlations with PT in PMN, which might provide novel information with regard to PCSK9 and hypercoagulability in PMN.
Collapse
Affiliation(s)
- Huizi Zhu
- Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Qian Meng
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Xiang Liu
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Chunjuan Zhai
- Department of Cardiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Jing Sun
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Rong Wang
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Liang Xu
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Xiaowei Yang
- Department of Nephrology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
5
|
Muso E, Kakita H, Suzuki H, Tsukamoto T. Updated evidence of beneficial effect of LDL apheresis for refractory nephrotic syndrome due to a variety of causative diseases for nationwide and global approval. Ther Apher Dial 2023; 27:987-999. [PMID: 37593995 DOI: 10.1111/1744-9987.14056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/23/2023] [Indexed: 08/19/2023]
Abstract
Low-density lipoprotein apheresis (LDL-A) therapy has shown reasonable efficacy in treating nephrotic syndrome (NS) refractory to initial drug therapy and has been covered by National Health Insurance for the indication of drug-resistant focal segmental glomerulosclerosis (FSGS) since 1992 in Japan and has contributed to liberating substantial number of patients of this disease from entering into end-stage renal disease by easier practical application in actual clinical settings. Subsequently, various beneficial evidence of this treatment has accumulated on those other than FSGS, however, due to the limitation of covered disease insurance only for FSGS, patients with diseases other than FSGS are unlikely to benefit from this treatment in practice. This review summarizes the therapeutic evidence of the beneficial effect of LDL-A accumulated to date and the mechanisms of action analyzed from multifaceted perspectives. examines the applicability of expanding insurance coverage for diseases other than FSGS.
Collapse
Affiliation(s)
- Eri Muso
- Department of Food and Nutrition, Faculty of Contemporary Home Economics, Kyoto Kacho University, Kyoto, Japan
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Hiroko Kakita
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Hiroyuki Suzuki
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Tatsuo Tsukamoto
- Department of Nephrology, Kitano Hospital, the Tazuke Kofukai Medical Research Institute, Osaka, Japan
| |
Collapse
|
6
|
Claudio P, Gabriella M. Nephrotic syndrome: pathophysiology and consequences. J Nephrol 2023; 36:2179-2190. [PMID: 37466816 DOI: 10.1007/s40620-023-01697-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 07/20/2023]
Abstract
In patients with kidney disease, nephrotic syndrome can lead to several complications including progressive kidney dysfunction. Proteinuria may lead to the formation of cellular or fibrous crescents with reciprocal development of rapidly progressive glomerulonephritis or focal glomerulosclerosis. Proteinuria may also cause overload and dysfunction of tubular epithelial cells, eventually resulting in tubular atrophy and interstitial fibrosis. Hypoalbuminemia is usually associated with increased risk of mortality and kidney dysfunction. Dyslipidemia may increase the risk of atherosclerotic complications, cause podocyte dysfunction and contribute to vascular thrombosis. Urinary loss of anticoagulants and overproduction of coagulation factors may facilitate a hypercoagulable state. Edema, hypogammaglobulinemia, loss of complement factors, and immunosuppressive therapy can favor infection. Treatment of these complications may reduce their impact on the severity of NS. Nephrotic syndrome is a kidney disorder that can worsen the quality of life and increase the risk of kidney disease progression.
Collapse
Affiliation(s)
| | - Moroni Gabriella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| |
Collapse
|
7
|
Skeby CK, Hummelgaard S, Gustafsen C, Petrillo F, Frederiksen KP, Olsen D, Kristensen T, Ivarsen P, Madsen P, Christensen EI, Nielsen R, Birn H, Glerup S, Weyer K. Proprotein convertase subtilisin/kexin type 9 targets megalin in the kidney proximal tubule and aggravates proteinuria in nephrotic syndrome. Kidney Int 2023; 104:754-768. [PMID: 37406929 DOI: 10.1016/j.kint.2023.06.024] [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: 10/06/2022] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023]
Abstract
Proteinuria is a prominent feature of chronic kidney disease. Interventions that reduce proteinuria slow the progression of chronic kidney disease and the associated risk of cardiovascular disease. Here, we propose a mechanistic coupling between proteinuria and proprotein convertase subtilisin/kexin type 9 (PCSK9), a regulator of cholesterol and a therapeutic target in cardiovascular disease. PCSK9 undergoes glomerular filtration and is captured by megalin, the receptor responsible for driving protein reabsorption in the proximal tubule. Accordingly, megalin-deficient mice and patients carrying megalin pathogenic variants (Donnai Barrow syndrome) were characterized by elevated urinary PCSK9 excretion. Interestingly, PCSK9 knockout mice displayed increased kidney megalin while PCSK9 overexpression resulted in its reduction. Furthermore, PCSK9 promoted trafficking of megalin to lysosomes in cultured proximal tubule cells, suggesting that PCSK9 is a negative regulator of megalin. This effect can be accelerated under disease conditions since either genetic destruction of the glomerular filtration barrier in podocin knockout mice or minimal change disease (a common cause of nephrotic syndrome) in patients resulted in enhanced tubular PCSK9 uptake and urinary PCSK9 excretion. Pharmacological PCSK9 inhibition increased kidney megalin while reducing urinary albumin excretion in nephrotic mice. Thus, glomerular damage increases filtration of PCSK9 and concomitantly megalin degradation, resulting in escalated proteinuria.
Collapse
Affiliation(s)
- Cecilie K Skeby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Camilla Gustafsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | | | | | - Ditte Olsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | - Tilde Kristensen
- Department of Internal Medicine, Renal Unit, Regional Hospital Viborg, Viborg, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Per Ivarsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Peder Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | | | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | - Kathrin Weyer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
8
|
Mitrofanova A, Merscher S, Fornoni A. Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease. Nat Rev Nephrol 2023; 19:629-645. [PMID: 37500941 DOI: 10.1038/s41581-023-00741-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Chronic kidney disease (CKD) is a global health problem with rising incidence and prevalence. Among several pathogenetic mechanisms responsible for disease progression, lipid accumulation in the kidney parenchyma might drive inflammation and fibrosis, as has been described in fatty liver diseases. Lipids and their metabolites have several important structural and functional roles, as they are constituents of cell and organelle membranes, serve as signalling molecules and are used for energy production. However, although lipids can be stored in lipid droplets to maintain lipid homeostasis, lipid accumulation can become pathogenic. Understanding the mechanisms linking kidney parenchymal lipid accumulation to CKD of metabolic or non-metabolic origin is challenging, owing to the tremendous variety of lipid species and their functional diversity across different parenchymal cells. Nonetheless, multiple research reports have begun to emphasize the effect of dysregulated kidney lipid metabolism in CKD progression. For example, altered cholesterol and fatty acid metabolism contribute to glomerular and tubular cell injury. Newly developed lipid-targeting agents are being tested in clinical trials in CKD, raising expectations for further therapeutic development in this field.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA.
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
9
|
Feng Z, Liao X, Peng J, Quan J, Zhang H, Huang Z, Yi B. PCSK9 causes inflammation and cGAS/STING pathway activation in diabetic nephropathy. FASEB J 2023; 37:e23127. [PMID: 37561547 DOI: 10.1096/fj.202300342rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023]
Abstract
Our previous research revealed that an increase in PCSK9 is linked to aggravated inflammation in the kidneys of mice affected by a high-fat diet and streptozotocin (HFD/STZ) or in HGPA-induced HK-2 cells. Furthermore, the cGAS/STING pathway has been reported to be involved in diabetic nephropathy (DN). Therefore, in this study, we aimed to examine the correlation between the proinflammatory effect of PCSK9 and the cGAS/STING pathway in DN. We used PCSK9 mAbs to inhibit PCSK9 in vivo and PCSK9 siRNA in vitro and measured the inflammatory phenotype in HFD/STZ-treated mice or HGPA-induced HK-2 cells, and observed decreased blood urea nitrogen, creatinine, UACR, and kidney injury in response to the PCSK9 mAb in HFD/STZ-treated mice. Moreover, IL-1 β, MCP-1, and TNF-α levels were reduced by the PCSK9 mAb in vivo and PCSK9 siRNA in vitro. We observed increased mtDNA damage and activation of the cGAS-STING signaling pathway during DN, as well as the downstream targets p-TBK1, p-NF-κB p65, and IL-1β. In a further experiment with an HGPA-induced DN model in HK-2 cells, we revealed that mtDNA damage was increased, which led to the activation of the cGAS/STING system and its downstream targets. Notably, the cGAS-STING signaling pathway was inhibited by the PCSK9 mAb in vivo and PCSK9 siRNA in vitro. In addition, inhibition of STING with C-176 in HGPA-induced HK-2 cells markedly blocked inflammation. In conclusion, we report for the first time that PCSK9 triggers mitochondrial DNA damage and activates the cGAS-STING pathway in DN, which leads to a series of inflammation cascades. PCSK9-targeted intervention can effectively reduce DN inflammation and delay its progression. Moreover, the inhibition of STING significantly abrogated the inflammation triggered by HGPA in HK-2 cells.
Collapse
Affiliation(s)
- Zhicai Feng
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Xiangyu Liao
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Juan Peng
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Jingjing Quan
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Hao Zhang
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Zhijun Huang
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
- Center for Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yi
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| |
Collapse
|
10
|
Hummelgaard S, Vilstrup JP, Gustafsen C, Glerup S, Weyer K. Targeting PCSK9 to tackle cardiovascular disease. Pharmacol Ther 2023; 249:108480. [PMID: 37331523 DOI: 10.1016/j.pharmthera.2023.108480] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Lowering blood cholesterol levels efficiently reduces the risk of developing atherosclerotic cardiovascular disease (ASCVD), including coronary artery disease (CAD), which is the main cause of death worldwide. CAD is caused by plaque formation, comprising cholesterol deposits in the coronary arteries. Proprotein convertase subtilisin kexin/type 9 (PCSK9) was discovered in the early 2000s and later identified as a key regulator of cholesterol metabolism. PCSK9 induces lysosomal degradation of the low-density lipoprotein (LDL) receptor in the liver, which is responsible for clearing LDL-cholesterol (LDL-C) from the circulation. Accordingly, gain-of-function PCSK9 mutations are causative of familial hypercholesterolemia, a severe condition with extremely high plasma cholesterol levels and increased ASCVD risk, whereas loss-of-function PCSK9 mutations are associated with very low LDL-C levels and protection against CAD. Since the discovery of PCSK9, extensive investigations in developing PCSK9 targeting therapies have been performed. The combined delineation of clear biology, genetic risk variants, and PCSK9 crystal structures have been major drivers in developing antagonistic molecules. Today, two antibody-based PCSK9 inhibitors have successfully progressed to clinical application and shown to be effective in reducing cholesterol levels and mitigating the risk of ASCVD events, including myocardial infarction, stroke, and death, without any major adverse effects. A third siRNA-based inhibitor has been FDA-approved but awaits cardiovascular outcome data. In this review, we outline the PCSK9 biology, focusing on the structure and nonsynonymous mutations reported in the PCSK9 gene and elaborate on PCSK9-lowering strategies under development. Finally, we discuss future perspectives with PCSK9 inhibition in other severe disorders beyond cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | - Kathrin Weyer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
11
|
Suzuki T, Iyoda M, Kanazawa N, Tachibana S, Honda H. Effect of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibition on Podocytes in Mouse Nephrotic Syndrome. J Transl Med 2023; 103:100199. [PMID: 37331628 DOI: 10.1016/j.labinv.2023.100199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is known to play a crucial role in dyslipidemia, and an increase in serum PCSK9 levels has also been reported in patients with nephrotic syndrome (NS). However, the specific effects of PCSK9 in kidney disease and the therapeutic potential of targeting PCSK9 in NS remain elusive. We thus investigated the effects of evolocumab (EVO) in mice with adriamycin (ADR)-induced NS. Male BALB/c mice were divided into the following 4 groups: Control, N = 11; EVO (monoclonal antibody for PCSK9), N = 11; ADR, N = 11; and ADR+EVO, N = 11. We also performed in vitro experiments using immortalized murine podocyte cells to validate the direct effects of PCSK9 on podocytes. EVO decreased urinary albumin levels and ameliorated podocytopathy in mice with ADR nephropathy. Further, EVO suppressed the Nod-like receptor protein 3 (NLRP3) inflammasome pathway in podocytes. PCSK9 expression upregulated CD36, a scavenger receptor of oxidized low-density lipoprotein (Ox-LDL), which in turn stimulated the absorption of Ox-LDL in vitro. EVO downregulated CD36 expression in podocytes both in vitro and in vivo. Immunofluorescence staining analysis reveals that CD36 and PCSK9 colocalized in the glomerular tufts of mice with ADR nephropathy. In the patients with focal segmental glomerulosclerosis, the CD36+ area in glomerular tufts increased compared with those diagnosed with minor glomerular abnormalities. This study revealed that EVO ameliorated mouse ADR nephropathy through the regulation of CD36 and NLRP3 inflammasome signaling. EVO treatment represents a potential therapeutic strategy for human NS.
Collapse
Affiliation(s)
- Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shohei Tachibana
- Department of Nephrology, Omiya Central General Hospital, Saitama, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Şener YZ, Tokgözoğlu L. Pleiotropy of PCSK9: Functions in Extrahepatic Tissues. Curr Cardiol Rep 2023; 25:979-985. [PMID: 37428313 DOI: 10.1007/s11886-023-01918-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE OF REVIEW Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a central role in the metabolism of LDL receptors and mainly acts in the liver. However, there are accumulating data that PCSK9 involves in several functions in different organs beyond the liver. Herein we aimed to summarize the effects of PCSK9 in tissues other than the liver. RECENT FINDINGS PCSK9 has crucial roles in heart, brain and kidney in addition to the cholesterol metabolism. Targeting PCSK9 for the treatment of hypercholesterolemia is effective in the prevention from cardiovascular diseases and PCSK9 inhibitors are getting to be administered in more cases. Therefore understanding the effects of PCSK9 in other tissues gained importance in the use of PCSK9 inhibitors era. PCSK9 participates in cardiac, renal, and neurologic functions however, current literature reveals that use of PSCSK9 inhibitors have beneficial or neutral effects on these organs. Inhibition of PCSK9 is assigned to be associated with new onset diabetes in experimental studies whereas real world data with PCSK9 inhibitors established no relationship between PCSK9 inhibitors and new onset diabetes. PCSK9 might be used as a target for the treatment of nephrotic syndrome and heart failure in the future.
Collapse
Affiliation(s)
- Yusuf Ziya Şener
- Cardiology Department, Beypazarı State Hospital, Ankara, Turkey.
| | - Lale Tokgözoğlu
- Cardiology Department, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
13
|
Wang D, Zhang D. Safety Issues Associated With the Clinical Application of PCSK9 Inhibitors: Current Findings. Cardiol Rev 2023; 31:155-161. [PMID: 35175957 DOI: 10.1097/crd.0000000000000441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Reduction in low-density lipoprotein cholesterol levels is the cornerstone of treatment and prevention of atherosclerotic cardiovascular diseases. Currently, high-intensity statins are being used as the first-line therapy to lower low-density lipoprotein cholesterol levels, as they improve the prognosis of patients with atherosclerotic cardiovascular disease and those in high-risk groups. However, in some patients, the expected reduction in cholesterol is not achieved despite aggressive treatment with statins. Moreover, some patients cannot tolerate the dosage or show poor response or compliance to statins. Therefore, combination therapies with statins and other medications should be considered. Recently, several clinical trials have shown that the use of proprotein convertase subtilisin/kexin type 9 inhibitors with or without statins and/or other lipid-lowering drugs can significantly reduce low-density lipoprotein cholesterol levels, sometimes to extremely low levels. Therefore, to facilitate appropriate prescription of these new lipid-lowering drugs, we systemically evaluated the safety issues associated with these inhibitors and extremely low low-density lipoprotein cholesterol levels.
Collapse
Affiliation(s)
- Dan Wang
- From the Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | | |
Collapse
|
14
|
Njeim R, Alkhansa S, Fornoni A. Unraveling the Crosstalk between Lipids and NADPH Oxidases in Diabetic Kidney Disease. Pharmaceutics 2023; 15:pharmaceutics15051360. [PMID: 37242602 DOI: 10.3390/pharmaceutics15051360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious complication of diabetes mellitus and a leading cause of end-stage renal disease. Abnormal lipid metabolism and intrarenal accumulation of lipids have been shown to be strongly correlated with the development and progression of diabetic kidney disease (DKD). Cholesterol, phospholipids, triglycerides, fatty acids, and sphingolipids are among the lipids that are altered in DKD, and their renal accumulation has been linked to the pathogenesis of the disease. In addition, NADPH oxidase-induced production of reactive oxygen species (ROS) plays a critical role in the development of DKD. Several types of lipids have been found to be tightly linked to NADPH oxidase-induced ROS production. This review aims to explore the interplay between lipids and NADPH oxidases in order to provide new insights into the pathogenesis of DKD and identify more effective targeted therapies for the disease.
Collapse
Affiliation(s)
- Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sahar Alkhansa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
15
|
Bell AS, Wagner J, Rosoff DB, Lohoff FW. Proprotein convertase subtilisin/kexin type 9 (PCSK9) in the central nervous system. Neurosci Biobehav Rev 2023; 149:105155. [PMID: 37019248 DOI: 10.1016/j.neubiorev.2023.105155] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023]
Abstract
The gene encoding proprotein convertase subtilisin/kexin type 9 (PCSK9) and its protein product have been widely studied for their role in cholesterol and lipid metabolism. PCSK9 increases the rate of metabolic degradation of low-density lipoprotein receptors, preventing the diffusion of low-density lipoprotein (LDL) from plasma into cells and contributes to high lipoprotein-bound cholesterol levels in the plasma. While most research has focused on the regulation and disease relevance of PCSK9 to the cardiovascular system and lipid metabolism, there is a growing body of evidence that PCSK9 plays a crucial role in pathogenic processes in other organ systems, including the central nervous system. PCSK9's impact on the brain is not yet fully understood, though several recent studies have sought to illuminate its impact on various neurodegenerative and psychiatric disorders, as well as its connection with ischemic stroke. Cerebral PCSK9 expression is low but is highly upregulated during disease states. Among others, PCSK9 is known to play a role in neurogenesis, neural cell differentiation, central LDL receptor metabolism, neural cell apoptosis, neuroinflammation, Alzheimer's Disease, Alcohol Use Disorder, and stroke. The PCSK9 gene contains several polymorphisms, including both gain-of-function and loss-of-function mutations which profoundly impact normal PCSK9 signaling and cholesterol metabolism. Gain-of-function mutations lead to persistent hypercholesterolemia and poor health outcomes, while loss-of-function mutations generally lead to hypocholesterolemia and may serve as a protective factor against diseases of the liver, cardiovascular system, and central nervous system. Recent genomic studies have sought to identify the end-organ effects of such mutations and continue to identify evidence of a much broader role for PCSK9 in extrahepatic organ systems. Despite this, there remain large gaps in our understanding of PCSK9, its regulation, and its effects on disease risk outside the liver. This review, which incorporates data from a wide range of scientific disciplines and experimental paradigms, is intended to describe PCSK9's role in the central nervous system as it relates to cerebral disease and neuropsychiatric disorders, and to examine the clinical potential of PCSK9 inhibitors and genetic variation in the PCSK9 gene on disease outcomes, including neurological and neuropsychiatric disease.
Collapse
|
16
|
Injection of an improperly stored proprotein convertase subtilisin/kexin type 9 monoclonal antibody in a patient with secondary dyslipidemia from nephrotic syndrome: a case report. J Med Case Rep 2023; 17:89. [PMID: 36899379 PMCID: PMC10007758 DOI: 10.1186/s13256-023-03804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/03/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Elevated plasma cholesterol and/or plasma triglyceride levels in nephrotic syndrome patients are the result of impaired lipoprotein clearance and a compensatory increase in hepatic lipoprotein synthesis. Plasma proprotein convertase subtilisin/kexin type 9 levels directly correlate to the amount of proteinuria in nephrotic syndrome patients. Proprotein convertase subtilisin/kexin type 9 monoclonal antibody has been used to treat dyslipidemia in some refractory nephrotic syndrome cases. As a therapeutic protein, proprotein convertase subtilisin/kexin type 9 monoclonal antibody simply deteriorates if stored in inappropriate temperatures or conditions. CASE PRESENTATION In this article, we present the case of a 16-year-old Thai female with severe combined dyslipidemia secondary to refractory nephrotic syndrome. She received proprotein convertase subtilisin/kexin type 9 monoclonal antibody (alirocumab) treatment. However, the drugs were mistakenly frozen in a freezer for up to 17 hours before being stored at 4 °C. After using two frozen devices, serum total cholesterol, free proprotein convertase subtilisin/kexin type 9, and lipoprotein(a) significantly decreased. Nonetheless, the patient developed a skin rash 2 weeks after the second injection and the lesion spontaneously resolved without any treatment approximately 1 month later. CONCLUSIONS The effectiveness of proprotein convertase subtilisin/kexin type 9 monoclonal antibody seems to be stable after being stored under freeze-thaw conditions. However, improperly stored drugs should be discarded to avoid any potential undesirable side effects.
Collapse
|
17
|
Lai X, Cui Z, Zhang H, Zhang YM, Wang F, Wang X, Meng LQ, Cheng XY, Liu G, Zhao MH. Long-term visit-to-visit variability in low-density lipoprotein cholesterol is associated with poor cardiovascular and kidney outcomes in patients with primary nephrotic syndrome. Int Urol Nephrol 2023; 55:1565-1574. [PMID: 36648742 DOI: 10.1007/s11255-023-03467-7] [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: 01/28/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
PURPOSE It is unclear whether long-term variability in low-density lipoprotein cholesterol (LDL-C) is associated with clinical outcomes in patients with nephrotic syndrome (NS). METHODS A large cohort of 1100 patients with primary NS underwent treatment and regular follow-up. Long-term variability in LDL-C was assessed by calculating its weighted standard deviation (w-SD). The primary endpoints of this study were the occurrence of arteriosclerotic cardiovascular disease (ASCVD) or kidney dysfunction. Factors associated with the w-SD of LDL-C were evaluated by linear regression. Associations of the w-SD of LDL-C with clinical outcomes were evaluated by Cox proportional hazards regression. RESULTS Over a median follow-up of 44.8 (interquartile range, 26.8, 70.1) months, 198 patients developed ASCVD (45.9 cases per 1,000 patient-years), and 84 patients developed kidney dysfunction (17.6 cases per 1,000 patient-years). The incidence rates of the primary outcomes increased across the quartiles of the w-SD of LDL-C (log-rank, P < 0.001). Multivariate Cox regression analysis showed that higher LDL-C variability was associated with an increased risk of ASCVD [hazard ratio (HR), 2.236; 95% confidence interval (CI), 1.684-2.969, P < 0.001] and an increased risk of kidney dysfunction (HR, 3.047; 95% CI 2.240-4.144, P < 0.001). The results were similar after adjusting the w-SD of LDL-C by its related parameters (baseline and mean LDL-C as well as mean total cholesterol), although the mean LDL-C was also an independent risk factor for ASCVD and kidney dysfunction. CONCLUSION Long-term variability in LDL-C was independently associated with the risk of ASCVD and kidney dysfunction in NS patients.
Collapse
Affiliation(s)
- Xuan Lai
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China.,Geriatrics Department, Peking University Third Hospital, Beijing, China
| | - Zhao Cui
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China.
| | - Hua Zhang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Yi-Miao Zhang
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Fang Wang
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Xin Wang
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Li-Qiang Meng
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Xu-Yang Cheng
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Gang Liu
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Institute of Nephrology, Key Laboratory of Renal Disease, Key Laboratory of CKD Prevention and Treatment, Peking University First Hospital, Peking University, Ministry of Health of China, Ministry of Education of China, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
18
|
Shinnawy HE, Fahmy AM, Gharib MS. Does pathological type of primary nephrotic syndrome affect serum concentrations of proprotein convertase subtilisin/kexin type 9? THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2022. [DOI: 10.1186/s43162-022-00157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Dyslipidemia is a common finding in primary nephrotic syndrome (PNS). Serum PCSK9 level is also increased in PNS and is the main cause of dyslipidemia in such patients. There is a paucity of data on the relation between dyslipidemia and pathological types of PNS. We hypothesized that severity of dyslipidemia varies across different types of PNS, and this variation is due to differences in serum PCSK9 levels.
Methods
Fifty patients recently diagnosed with PNS were included in this cross-sectional study. Serum PCSK9, albumin, creatinine, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglycerides (TG), and 24-h urine protein were measured. Comparison of patients’ characteristics between pathological types of PNS and correlations between serum PCSK9 and other parameters were assessed.
Results
Serum PCSK9 levels were significantly higher in PNS patients compared with healthy individuals (314.58 ± 73.83 vs 253.42 ± 36.66 ng/ml, p < 0.001). No differences found between PNS types regarding serum levels of PCSK9 (p = 0.571), TC (p = 0.806), LDL-C (p = 0.950), HDL-C (p = 0.844), VLDL-C (p = 0.472), and TG (p = 0.969). Serum PCSK9 levels correlated significantly with TC (p < 0.001), LDL-C (p < 0.001), HDL-C (p = 0.003), VLDL-C (p = 0.008), TG (p = 0.005), 24-h urine protein (p = 0.005), and male sex (p = 0.014).
Conclusion
The pathological type of PNS does not affect serum levels of PCSK9 and components of lipid profile.
Collapse
|
19
|
Zuzda K, Grycuk W, Małyszko J, Małyszko J. Kidney and lipids: novel potential therapeutic targets for dyslipidemia in kidney disease? Expert Opin Ther Targets 2022; 26:995-1009. [PMID: 36548906 DOI: 10.1080/14728222.2022.2161887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Altered lipid distribution and metabolism may lead to the development and/or progression of chronic kidney disease (CKD). Dyslipidemia is a major risk factor for CKD and increases the risk of cardiovascular events and mortality. Therefore, lipid-lowering treatments may decrease cardiovascular risk and prevent death. AREAS COVERED Key players involved in regulating lipid accumulation in the kidney; contribution of lipids to CKD progression, lipotoxicity, and mitochondrial dysfunction in kidney disease; recent therapeutic approaches for dyslipidemia. EXPERT OPINION The precise mechanisms for regulating lipid metabolism, particularly in kidney disease, are poorly understood. Guidelines for lipid-lowering therapy for CKD are controversial. Several hypolipemic therapies are available, but compared to others, statin therapy is the most common. No clinical trial has evaluated the efficacy of proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) in preventing cardiovascular events or improving kidney function among patients with CKD or kidney transplant recipients. Attractive alternatives, such as PCSK9-small interfering RNA (siRNA) molecules or evinacumab are available. Additionally, several promising agents, such as cyclodextrins and the FXR/TGR5 dual agonist, INT-767, can improve renal lipid metabolism disorders and delay CKD progression. Drugs targeting mitochondrial dysfunction could be an option for the treatment of dyslipidemia and lipotoxicity, particularly in renal diseases.
Collapse
Affiliation(s)
- Konrad Zuzda
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Bialystok, Poland
| | - Wiktoria Grycuk
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Bialystok, Poland
| | - Jacek Małyszko
- 1st Department of Nephrology and Transplantology, Medical University of Bialystok, Bialystok, Poland
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Bialystok, Poland
| |
Collapse
|
20
|
The role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in the pathophysiology of psoriasis and systemic lupus erythematosus. Postepy Dermatol Alergol 2022; 39:645-650. [PMID: 36090718 PMCID: PMC9454343 DOI: 10.5114/ada.2022.118919] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/01/2021] [Indexed: 11/29/2022] Open
Abstract
Inflammation and atherogenic dyslipidaemia are often observed in skin diseases and represent an increased risk of cardiovascular disorders. Proprotein convertase subtilisin/kexin type 9 plays an important role in the regulation of serum low-density lipoprotein cholesterol levels. Its biological role, however, seems to go much beyond the regulation of cholesterol metabolism. The article presents potential pathophysiological links between inflammatory process and lipid disorders based on the example of psoriasis and systemic lupus erythematosus.
Collapse
|
21
|
Doiron S, Paquette M, Baass A, Bollée G, Cardinal H, Bernard S. Association between circulating PCSK9 and proteinuria in nephrotic syndrome: A cross-sectional study. Clin Biochem 2022; 109-110:51-56. [DOI: 10.1016/j.clinbiochem.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/18/2022] [Accepted: 08/02/2022] [Indexed: 11/29/2022]
|
22
|
Xiao M, Bohnert BN, Grahammer F, Artunc F. Rodent models to study sodium retention in experimental nephrotic syndrome. Acta Physiol (Oxf) 2022; 235:e13844. [PMID: 35569011 DOI: 10.1111/apha.13844] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
Sodium retention and edema are hallmarks of nephrotic syndrome (NS). Different experimental rodent models have been established for simulating NS, however, not all of them feature sodium retention which requires proteinuria to exceed a certain threshold. In rats, puromycin aminonucleoside nephrosis (PAN) is a classic NS model introduced in 1955 that was adopted as doxorubicin-induced nephropathy (DIN) in 129S1/SvImJ mice. In recent years, mice with inducible podocin deletion (Nphs2Δipod ) or podocyte apoptosis (POD-ATTAC) have been developed. In these models, sodium retention is thought to be caused by activation of the epithelial sodium channel (ENaC) in the distal nephron through aberrantly filtered serine proteases or proteasuria. Strikingly, rodent NS models follow an identical chronological time course after the development of proteinuria featuring sodium retention within days and spontaneous reversal thereafter. In DIN and Nphs2Δipod mice, inhibition of ENaC by amiloride or urinary serine protease activity by aprotinin prevents sodium retention, opening up new and promising therapeutic approaches that could be translated into the treatment of nephrotic patients. However, the essential serine protease(s) responsible for ENaC activation is (are) still unknown. With the use of nephrotic rodent models, there is the possibility that this (these) will be identified in the future. This review summarizes the various rodent models used to study experimental nephrotic syndrome and the insights gained from these models with regard to the pathophysiology of sodium retention.
Collapse
Affiliation(s)
- Mengyun Xiao
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
| | - Bernhard N. Bohnert
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| | - Florian Grahammer
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Ferruh Artunc
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| |
Collapse
|
23
|
Byun JH, Lebeau PF, Platko K, Carlisle RE, Faiyaz M, Chen J, MacDonald ME, Makda Y, Yousof T, Lynn EG, Dickhout JG, Krepinsky JC, Weaver F, Igdoura SA, Seidah NG, Austin RC. Inhibitory Antibodies against PCSK9 Reduce Surface CD36 and Mitigate Diet-Induced Renal Lipotoxicity. KIDNEY360 2022; 3:1394-1410. [PMID: 36176646 PMCID: PMC9416829 DOI: 10.34067/kid.0007022021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/26/2022] [Indexed: 01/11/2023]
Abstract
Background PCSK9 modulates the uptake of circulating lipids through a range of receptors, including the low-density lipoprotein receptor (LDLR) and CD36. In the kidney, CD36 is known to contribute to renal injury through pro-inflammatory and -fibrotic pathways. In this study, we sought to investigate the role of PCSK9 in modulating renal lipid accumulation and injury through CD36 using a high fat diet (HFD)-induced murine model. Methods The effect of PCSK9 on the expression of CD36 and intracellular accumulation of lipid was examined in cultured renal cells and in the kidneys of male C57BL/6J mice. The effect of these findings was subsequently explored in a model of HFD-induced renal injury in Pcsk9 -/- and Pcsk9 +/+ littermate control mice on a C57BL/6J background. Results In the absence of PCSK9, we observed heightened CD36 expression levels, which increased free fatty acid (FFA) uptake in cultured renal tubular cells. As a result, PCSK9 deficiency was associated with an increase in long-chain saturated FFA-induced ER stress. Consistent with these observations, Pcsk9-/- mice fed a HFD displayed elevated ER stress, inflammation, fibrosis, and renal injury relative to HFD-fed control mice. In contrast to Pcsk9-/- mice, pretreatment of WT C57BL/6J mice with evolocumab, an anti-PCSK9 monoclonal antibody (mAb) that binds to and inhibits the function of circulating PCSK9, protected against HFD-induced renal injury in association with reducing cell surface CD36 expression on renal epithelia. Conclusions We report that circulating PCSK9 modulates renal lipid uptake in a manner dependent on renal CD36. In the context of increased dietary fat consumption, the absence of circulating PCSK9 may promote renal lipid accumulation and subsequent renal injury. However, although the administration of evolocumab blocks the interaction of PCSK9 with the LDLR, this evolocumab/PCSK9 complex can still bind CD36, thereby protecting against HFD-induced renal lipotoxicity.
Collapse
Affiliation(s)
- Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Paul F. Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Rachel E. Carlisle
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Mahi Faiyaz
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Jack Chen
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Melissa E. MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Yumna Makda
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Tamana Yousof
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Edward G. Lynn
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Jeffrey G. Dickhout
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Joan C. Krepinsky
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| | - Fiona Weaver
- Department of Biology and Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Suleiman A. Igdoura
- Department of Biology and Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Nabil G. Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, University of Montreal, Montreal, Canada
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe’s Hamilton and The Hamilton Centre for Kidney Research, Hamilton, Canada
| |
Collapse
|
24
|
Kheirkhah A, Lamina C, Kollerits B, Schachtl-Riess J, Schultheiss U, Forer L, Sekula P, Kotsis F, Eckardt KU, Kronenberg F. PCSK9 and Cardiovascular Disease in Individuals with Moderately Decreased Kidney Function. Clin J Am Soc Nephrol 2022; 17:809-818. [PMID: 35387881 PMCID: PMC9269648 DOI: 10.2215/cjn.01230122] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/01/2022] [Indexed: 11/23/2022]
Abstract
Background and objectives: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key regulator of lipid homeostasis. Studies investigating the association between PCSK9 and cardiovascular disease in large cohorts of CKD patients are limited. Design, setting, participants, and measurements: The association of PCSK9 concentrations with prevalent and incident cardiovascular disease was investigated in 5,138 Caucasians of the German Chronic Kidney Disease study with a median follow-up of 6.5 years. Inclusion criteria were eGFR of 30-60 mL/min/1.73 m2 or an eGFR>60 mL/min/1.73m2 in the presence of overt proteinuria (urine albumin-creatinine ratio >300mg/g or equivalent). Prevalent cardiovascular disease was defined as history of non-fatal myocardial infarction, coronary artery bypass grafting, percutaneous transluminal coronary angioplasty, carotid arteries interventions and stroke. Incident major adverse cardiovascular disease events included death from cardiovascular causes, acute non-fatal myocardial infarction and non-fatal stroke. Results: Median PCSK9 concentration in the cohort was 285 ng/mL (interquartile range 231-346 ng/mL). There was no association between PCSK9 concentrations and baseline eGFR and albuminuria. With each 100 ng/ml increment of PCSK9, the odds for prevalent cardiovascular disease (n=1,284) was 1.22-fold (95%CI 1.12-1.34, p<0.001) higher in a model with extended adjustment for major confounders. This association was stronger in non-statin than statin-users (p-value for interaction=0.009). During follow-up 474 individuals experienced a major adverse cardiovascular disease event and participants in PCSK9 quartiles 2 to 4 had a 32%-47% higher risk compared to quartile 1 (p<0.05). Subgroup analysis revealed this association was restricted to those participants who already had cardiovascular disease at baseline (all HR>1.75, p<0.05). In addition, PCSK9 showed a valuable gain in classification accuracy both for prevalent cardiovascular disease (NRI=0.27, 95%CI:0.20-0.33) and incident major adverse cardiovascular disease events during follow-up (NRI=0.10, 95%CI:0.008-0.21) when added to an extended adjustment model. Conclusions: Our findings reveal no relation of PCSK9 with baseline eGFR and albuminuria, but a significant association between higher PCSK9 concentrations and risk of cardiovascular disease independent of traditional risk factors including LDL-cholesterol levels.
Collapse
Affiliation(s)
- Azin Kheirkhah
- A Kheirkhah, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- C Lamina, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Kollerits
- B Kollerits, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna Schachtl-Riess
- J Schachtl-Riess, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulla Schultheiss
- U Schultheiss, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Lukas Forer
- L Forer, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Peggy Sekula
- P Sekula, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Fruzsina Kotsis
- F Kotsis, Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Kai-Uwe Eckardt
- K Eckardt, University Hospital Erlangen, Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Florian Kronenberg
- F Kronenberg, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Kajingulu FPM, Lepira FB, Nkodila AN, Makulo JRR, Mokoli VM, Ekulu PM, Bukabau JB, Nlandu YM, Longo AL, Nseka NM, Labriola L, Sumaili EK. Circulating Proprotein Convertase Subtilisin/Kexin type 9 level independently predicts incident cardiovascular events and all-cause mortality in hemodialysis black Africans patients. BMC Nephrol 2022; 23:123. [PMID: 35354429 PMCID: PMC8969257 DOI: 10.1186/s12882-022-02748-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/14/2022] [Indexed: 01/01/2023] Open
Abstract
Background Cardiovascular (CV) disease is the leading cause of mortality in patients with end-stage kidney disease (ESKD). The aim of the present study was to determine whether Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) could be an independent predictor of CV events and all-cause mortality in black African haemodialysis patients. Methods We carried out a prospective cohort study of all consecutive hemodialysis (HD) patients between August 2016 and July 2020, admitted in six hemodialysis centers of Kinshasa, Democratic Republic of Congo. Independent determinants of plasma PCSK-9 measured by ELISA were sought using multiple linear regression analysis. Kaplan-Meier’s method described the incidence of CV events while competitive and proportional risk models looked for independent risk factors for death at the .05 significance level. Results Out of 207 HD patients, 91 (43.9%) died; 116 (56.1%) have survived. PCSK9 level was significantly higher in deceased patients compared to survivors: 28.0 (24.0–31.0) ng/l vs 9.6 (8.6–11.6) ng/ml (p < 0.001). Patients with plasma PCSK9 levels in tertile 3 had a higher incidence of CV events and mortality compared to patients with plasma PCSK9 levels in tertile 2 or tertile 1 (p < 0.001). Tertile 3 negatively influence survival rates (26.6%) compared to tertile 2 (54.7%) and tertile 1 (85.3%). Patients in tertile 3 and tertile 2 had a 4-fold higher risk of death than patients in tertile 1. After adjustment for all parameters, competitive risk analysis showed that mortality was 2 times higher in patients with stroke. Similarly, serum albumin < 3.5 g/dL or PCSK9 in tertile 3 were respectively associated with 2 or 6 times higher rates of deaths. Conclusion Elevated plasma PCSK9 level is an independent major predictor of incident CV events and all-cause mortality in black African HD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02748-0.
Collapse
Affiliation(s)
- François-Pantaléon Musungayi Kajingulu
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo.
| | - François Bompeka Lepira
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Aliocha Natuhoyila Nkodila
- Faculty of Family Medicine and Primary care, University of Protestant in Congo, Kinshasa, Democratic Republic of Congo
| | - Jean-Robert Rissassy Makulo
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Vieux Momeme Mokoli
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Pepe Mfutu Ekulu
- Department of Pediatrics, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Justine Busanga Bukabau
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Yannick Mayamba Nlandu
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Augustin Luzayadio Longo
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Nazaire Mangani Nseka
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Laura Labriola
- Université Catholique de Louvain / Cliniques Universitaires Saint Luc de Bruxelles, Brussels, Belgium
| | - Ernest Kiswaya Sumaili
- Department of Internal Medicine, Division of Nephrology, Kinshasa University Hospital, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| |
Collapse
|
26
|
Maligłówka M, Kosowski M, Hachuła M, Cyrnek M, Bułdak Ł, Basiak M, Bołdys A, Machnik G, Bułdak RJ, Okopień B. Insight into the Evolving Role of PCSK9. Metabolites 2022; 12:metabo12030256. [PMID: 35323699 PMCID: PMC8951079 DOI: 10.3390/metabo12030256] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is the last discovered member of the family of proprotein convertases (PCs), mainly synthetized in hepatic cells. This serine protease plays a pivotal role in the reduction of the number of low-density lipoprotein receptors (LDLRs) on the surface of hepatocytes, which leads to an increase in the level of cholesterol in the blood. This mechanism and the fact that gain of function (GOF) mutations in PCSK9 are responsible for causing familial hypercholesterolemia whereas loss-of-function (LOF) mutations are associated with hypocholesterolemia, prompted the invention of drugs that block PCSK9 action. The high efficiency of PCSK9 inhibitors (e.g., alirocumab, evolocumab) in decreasing cardiovascular risk, pleiotropic effects of other lipid-lowering drugs (e.g., statins) and the multifunctional character of other proprotein convertases, were the cause for proceeding studies on functions of PCSK9 beyond cholesterol metabolism. In this article, we summarize the current knowledge on the roles that PCSK9 plays in different tissues and perspectives for its clinical use.
Collapse
Affiliation(s)
- Mateusz Maligłówka
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
- Correspondence:
| | - Michał Kosowski
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Hachuła
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Cyrnek
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Marcin Basiak
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Aleksandra Bołdys
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| | - Rafał Jakub Bułdak
- Institute of Medical Sciences, University of Opole, 45-040 Opole, Poland;
| | - Bogusław Okopień
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia in Katowice, 40-007 Katowice, Poland; (M.K.); (M.H.); (M.C.); (Ł.B.); (M.B.); (A.B.); (G.M.); (B.O.)
| |
Collapse
|
27
|
Semova I, Levenson AE, Krawczyk J, Bullock K, Gearing ME, Ling AV, Williams KA, Miao J, Adamson SS, Shin DJ, Chahar S, Graham MJ, Crooke RM, Hagey LR, Vicent D, de Ferranti SD, Kidambi S, Clish CB, Biddinger SB. Insulin Prevents Hypercholesterolemia by Suppressing 12a-Hydroxylated Bile Acid Production. Circulation 2022; 145:969-982. [PMID: 35193378 PMCID: PMC9365453 DOI: 10.1161/circulationaha.120.045373] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The risk of cardiovascular disease in type 1 diabetes remains extremely high, despite marked advances in blood glucose control and even the widespread use of cholesterol synthesis inhibitors. Thus, a deeper understanding of insulin regulation of cholesterol metabolism, and its disruption in type 1 diabetes, could reveal better treatment strategies. Methods: To define the mechanisms by which insulin controls plasma cholesterol levels, we knocked down the insulin receptor, FoxO1, and the key bile acid synthesis enzyme, CYP8B1. We measured bile acid composition, cholesterol absorption, and plasma cholesterol. In parallel, we measured markers of cholesterol absorption and synthesis in humans with type 1 diabetes treated with ezetimibe and statins in a double-blind crossover study. Results: Mice with hepatic deletion of the insulin receptor showed marked increases in 12α-hydroxylated bile acids (12HBAs), cholesterol absorption, and plasma cholesterol. This phenotype was entirely reversed by hepatic deletion of FoxO1. FoxO1 is inhibited by insulin, and required for the production of 12HBAs, which promote intestinal cholesterol absorption and suppress hepatic cholesterol synthesis. Knockdown of Cyp8b1 normalized 12HBA levels and completely prevented hypercholesterolemia in mice with hepatic deletion of the insulin receptor (n=5-30) as well as mouse models of type 1 diabetes (n=5-22). In parallel, the cholesterol absorption inhibitor, ezetimibe, normalized cholesterol absorption and LDL-cholesterol in patients with type 1 diabetes as well as, or better than, the cholesterol synthesis inhibitor, simvastatin (n=20). Conclusions: Insulin, by inhibiting FoxO1 in the liver, reduces 12HBAs, cholesterol absorption, and plasma cholesterol levels. Thus, type 1 diabetes leads to a unique set of derangements in cholesterol metabolism, with increased absorption rather than synthesis. These derangements are reversed by ezetimibe, but not statins, which are currently the first line of lipid-lowering treatment in type 1 diabetes. Taken together, these data suggest that a personalized approach to lipid lowering in type 1 diabetes may be more effective and highlight the need for further studies specifically in this group of patients.
Collapse
Affiliation(s)
- Ivana Semova
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Amy E Levenson
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Joanna Krawczyk
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | - Mary E Gearing
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Alisha V Ling
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Kathryn A Williams
- Biostatistics and Research Design Center, ICCTR, Boston Children's Hospital, Boston, MA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Stuart S Adamson
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Dong-Ju Shin
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Satyapal Chahar
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | | | - Lee R Hagey
- Department of Medicine, University of California, San Diego, CA
| | - David Vicent
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Sarah D de Ferranti
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Srividya Kidambi
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | | | - Sudha B Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
PCSK9 promotes arterial medial calcification. Atherosclerosis 2022; 346:86-97. [DOI: 10.1016/j.atherosclerosis.2022.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/18/2022]
|
29
|
Guo Y, Tang Z, Yan B, Yin H, Tai S, Peng J, Cui Y, Gui Y, Belke D, Zhou S, Zheng XL. PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Triggers Vascular Smooth Muscle Cell Senescence and Apoptosis: Implication of Its Direct Role in Degenerative Vascular Disease. Arterioscler Thromb Vasc Biol 2021; 42:67-86. [PMID: 34809446 DOI: 10.1161/atvbaha.121.316902] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE PCSK9 (proprotein convertase subtilisin/kexin type 9) plays a critical role in cholesterol metabolism via the PCSK9-LDLR (low-density lipoprotein receptor) axis in the liver; however, evidence indicates that PCSK9 directly contributes to the pathogenesis of various diseases through mechanisms independent of its LDL-cholesterol regulation. The objective of this study was to determine how PCSK9 directly acts on vascular smooth muscle cells (SMCs), contributing to degenerative vascular disease. Approach and Results: We first examined the effects of PCSK9 on cultured human aortic SMCs. Overexpression of PCSK9 downregulated the expression of ApoER2 (apolipoprotein E receptor 2), a known target of PCSK9. Treatment with soluble recombinant human ApoER2 or the DNA synthesis inhibitor, hydroxyurea, inhibited PCSK9-induced polyploidization and other cellular responses of human SMCs. Treatment with antibodies against ApoER2 resulted in similar effects to those observed with PCSK9 overexpression. Inducible, SMC-specific knockout of Pcsk9 accelerated neointima formation in mouse carotid arteries and reduced age-related arterial stiffness. PCSK9 was expressed in SMCs of human atherosclerotic lesions and abundant in the "shoulder" regions of vulnerable atherosclerotic plaques. PCSK9 was also expressed in SMCs of abdominal aortic aneurysm, which was inversely related to the expression of smooth muscle α-actin. CONCLUSIONS Our findings demonstrate that PCSK9 inhibits proliferation and induces polyploidization, senescence, and apoptosis, which may be relevant to various degenerative vascular diseases.
Collapse
Affiliation(s)
- Yanan Guo
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Zhihan Tang
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Binjie Yan
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Hao Yin
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Now with Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada (H.Y.)
| | - Shi Tai
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yuting Cui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yu Gui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Darrell Belke
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Shenghua Zhou
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| |
Collapse
|
30
|
Use of Lipid-Modifying Agents for the Treatment of Glomerular Diseases. J Pers Med 2021; 11:jpm11080820. [PMID: 34442464 PMCID: PMC8401447 DOI: 10.3390/jpm11080820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Although dyslipidemia is associated with chronic kidney disease (CKD), it is more common in nephrotic syndrome (NS), and guidelines for the management of hyperlipidemia in NS are largely opinion-based. In addition to the role of circulating lipids, an increasing number of studies suggest that intrarenal lipids contribute to the progression of glomerular diseases, indicating that proteinuric kidney diseases may be a form of "fatty kidney disease" and that reducing intracellular lipids could represent a new therapeutic approach to slow the progression of CKD. In this review, we summarize recent progress made in the utilization of lipid-modifying agents to lower renal parenchymal lipid accumulation and to prevent or reduce kidney injury. The agents mentioned in this review are categorized according to their specific targets, but they may also regulate other lipid-relevant pathways.
Collapse
|
31
|
Maligłówka M, Bułdak Ł, Okopień B, Bołdys A. The consequences of PCSK9 inhibition in selected tissues. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.9127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is one of nine members of the proprotein
convertase family. These serine proteases play a pivotal role in the post-translational
modification of proteins and the activation of hormones, enzymes, transcription factors and
growth factors. As a result, they participate in many physiological processes like embryogenesis,
activity of central nervous system and lipid metabolism. Scientific studies show
that the family of convertases is also involved in the pathogenesis of viral and bacterial
infections, osteoporosis, hyperglycaemia, cardiovascular diseases, neurodegenerative disorders
and cancer. The inhibition of PCSK9 by two currently approved for use monoclonal
antibodies (alirocumab, evolocumab) slows down the degradation of low-density lipoprotein
cholesterol receptors (LDLRs). This leads to increased density of LDLRs on the surface
of hepatocytes, resulting in decreased level of low-density lipoprotein cholesterol (LDL-C)
in the bloodstream, which is connected with the reduction of cardiovascular risk. PCSK9 inhibitors (PCSK9i) were created for the patients who could not achieve appropriate level
of LDL-C using current statin and ezetimibe therapy. It seems that high therapeutic efficacy
of PCSK9i will make them more common in the clinical use. The pleiotropic effects
of previously mentioned lipid-lowering therapies were the reasons for literature review of
possible positive and negative effects of PCSK9 inhibition beyond cholesterol metabolism.
Collapse
Affiliation(s)
- Mateusz Maligłówka
- Katedra Farmakologii, Klinika Chorób Wewnętrznych i Farmakologii Klinicznej, Wydział Nauk Medycznych Śląskiego Uniwersytetu Medycznego w Katowicach
| | - Łukasz Bułdak
- Katedra Farmakologii, Klinika Chorób Wewnętrznych i Farmakologii Klinicznej, Wydział Nauk Medycznych Śląskiego Uniwersytetu Medycznego w Katowicach
| | - Bogusław Okopień
- Katedra Farmakologii, Klinika Chorób Wewnętrznych i Farmakologii Klinicznej, Wydział Nauk Medycznych Śląskiego Uniwersytetu Medycznego w Katowicach
| | - Aleksandra Bołdys
- Katedra Farmakologii, Klinika Chorób Wewnętrznych i Farmakologii Klinicznej, Wydział Nauk Medycznych Śląskiego Uniwersytetu Medycznego w Katowicach
| |
Collapse
|
32
|
Shrestha P, Adepu S, Vivès RR, Masri RE, Klooster A, Kaptein F, Dam W, Bakker SJL, van Goor H, van de Sluis B, van den Born J. Hypercholesterolemia in Progressive Renal Failure Is Associated with Changes in Hepatic Heparan Sulfate - PCSK9 Interaction. J Am Soc Nephrol 2021; 32:1371-1388. [PMID: 33758009 PMCID: PMC8259657 DOI: 10.1681/asn.2020091376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 02/04/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Dyslipidemia is an important risk factor in CKD. The liver clears triglyceride-rich lipoproteins (TRL) via LDL receptor (LDLR), LDLR-related protein-1 (LRP-1), and heparan sulfate proteoglycans (HSPGs), mostly syndecan-1. HSPGs also facilitate LDLR degradation by proprotein convertase subtilisin/kexin type 9 (PCSK9). Progressive renal failure affects the structure and activity of hepatic lipoprotein receptors, PCSK9, and plasma cholesterol. METHODS Uninephrectomy- and aging-induced CKD in normotensive Wistar rats and hypertensive Munich-Wistar-Frömter (MWF) rats. RESULTS Compared with 22-week-old sex- and strain-matched rats, 48-week-old uninephrectomized Wistar-CKD and MWF-CKD rats showed proteinuria, increased plasma creatinine, and hypercholesterolemia (all P<0.05), which were most apparent in hypertensive MWF-CKD rats. Hepatic PCSK9 expression increased in both CKD groups (P<0.05), with unusual sinusoidal localization, which was not seen in 22-week-old rats. Heparan sulfate (HS) disaccharide analysis, staining with anti-HS mAbs, and mRNA expression of HS polymerase exostosin-1 (Ext-1), revealed elongated HS chains in both CKD groups. Solid-phase competition assays showed that the PCSK9 interaction with heparin-albumin (HS-proteoglycan analogue) was critically dependent on polysaccharide chain length. VLDL binding to HS from CKD livers was reduced (P<0.05). Proteinuria and plasma creatinine strongly associated with plasma cholesterol, PCSK9, and HS changes. CONCLUSIONS Progressive CKD induces hepatic HS elongation, leading to increased interaction with PCSK9. This might reduce hepatic lipoprotein uptake and thereby induce dyslipidemia in CKD. Therefore, PCSK9/HS may be a novel target to control dyslipidemia.
Collapse
Affiliation(s)
- Pragyi Shrestha
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Saritha Adepu
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Romain R. Vivès
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Rana El Masri
- University Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Energie Atomique (CEA), Institute of Structural Biology, Grenoble, France
| | - Astrid Klooster
- Department of Pathology, Pathology Friesland, Leeuwarden, The Netherlands
| | - Fleur Kaptein
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wendy Dam
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J. L. Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department Pediatrics, Section of Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
33
|
Song EJ, Ahn S, Min SK, Ha J, Oh GT. Combined application of rapamycin and atorvastatin improves lipid metabolism in apolipoprotein E-deficient mice with chronic kidney disease. BMB Rep 2021. [PMID: 33050984 PMCID: PMC8016660 DOI: 10.5483/bmbrep.2021.54.3.136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis arising from the pro-inflammatory conditions associated with chronic kidney disease (CKD) increases major cardiovascular morbidity and mortality. Rapamycin (RAPA) is known to inhibit atherosclerosis under CKD and non-CKD conditions, but it can cause dyslipidemia; thus, the co-application of lipid-lowering agents is recommended. Atorvastatin (ATV) has been widely used to reduce serum lipids levels, but its synergistic effect with RAPA in CKD remains unclear. Here, we analyzed the effect of their combined treatment on atherosclerosis stimulated by CKD in apolipoprotein E-deficient (ApoE−/−) mice. Oil Red O staining revealed that treatment with RAPA and RAPA+ ATV, but not ATV alone, significantly decreased the atherosclerotic lesions in the aorta and aortic sinus, compared to those seen in the control (CKD) group. The co-administration of RAPA and ATV improved the serum lipid profile and raised the expression levels of proteins involved in reverse cholesterol transport (LXRα, CYP7A1, ABCG1, PPARγ, ApoA1) in the liver. The CKD group showed increased levels of various genes encoding atherosclerosis-promoting cytokines in the spleen (Tnf-α, Il-6 and Il-1β) and aorta (Tnf-α and Il-4), and these increases were attenuated by RAPA treatment. ATV and RAPA+ATV decreased the levels of Tnf-α and Il-1β in the spleen, but not in the aorta. Together, these results indicate that, in CKD-induced ApoE−/− mice, RAPA significantly reduces the development of atherosclerosis by regulating the expression of inflammatory cytokines and the co-application of ATV improves lipid metabolism.
Collapse
Affiliation(s)
- Eun Ju Song
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
- Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Sanghyun Ahn
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung-Kee Min
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
34
|
Kim JJ, Wilbon SS, Fornoni A. Podocyte Lipotoxicity in CKD. KIDNEY360 2021; 2:755-762. [PMID: 35373048 PMCID: PMC8791311 DOI: 10.34067/kid.0006152020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/24/2021] [Indexed: 02/06/2023]
Abstract
CKD represents the ninth most common cause of death in the United States but, despite this large health burden, treatment options for affected patients remain limited. To remedy this, several relevant pathways have been identified that may lead to novel therapeutic options. Among them, altered renal lipid metabolism, first described in 1982, has been recognized as a common pathway in clinical and experimental CKD of both metabolic and nonmetabolic origin. This observation has led many researchers to investigate the cause of this renal parenchyma lipid accumulation and its downstream effect on renal structure and function. Among key cellular components of the kidney parenchyma, podocytes are terminally differentiated cells that cannot be easily replaced when lost. Clinical and experimental evidence supports a role of reduced podocyte number in the progression of CKD. Given the importance of the podocytes in the maintenance of the glomerular filtration barrier and the accumulation of TG and cholesterol-rich lipid droplets in the podocyte and glomerulus in kidney diseases that cause CKD, understanding the upstream cause and downstream consequences of lipid accumulation in podocytes may lead to novel therapeutic opportunities. In this review, we hope to consolidate our understanding of the causes and consequences of dysregulated renal lipid metabolism in CKD development and progression, with a major focus on podocytes.
Collapse
|
35
|
Shrestha P, Yazdani S, Vivès RR, El Masri R, Dam W, van de Sluis B, van den Born J. Proteinuria converts hepatic heparan sulfate to an effective proprotein convertase subtilisin kexin type 9 enzyme binding partner. Kidney Int 2021; 99:1369-1381. [PMID: 33609572 DOI: 10.1016/j.kint.2021.01.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/06/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022]
Abstract
Hepatic uptake of triglyceride-rich remnant lipoproteins is mediated by the low-density lipoprotein receptor, a low-density lipoprotein receptor related protein and the heparan sulfate proteoglycan, syndecan-1. Heparan sulfate proteoglycan also mediates low-density lipoprotein receptor degradation by a regulator of cholesterol homeostasis, proprotein convertase subtilisin kexin type 9 (PCSK9), thereby hampering triglyceride-rich remnant lipoproteins uptake. In this study, we investigated the effects of proteinuria on PCSK9, hepatic heparan sulfate proteoglycan and plasma triglyceride-rich remnant lipoproteins. Adriamycin-injected rats developed proteinuria, elevated triglycerides and total cholesterol (all significantly increased). Proteinuria associated with triglycerides and total cholesterol and serum PCSK9 (all significant associations) without loss of the low-density lipoprotein receptor as evidenced by immunofluorescence staining and western blotting. In proteinuric rats, PCSK9 accumulated in sinusoids, whereas in control rats PCSK9 was localized in the cytoplasm of hepatocytes. Molecular profiling revealed that the heparan sulfate side chains of heparan sulfate proteoglycan to be hypersulfated in proteinuric rats. Competition assays revealed sulfation to be a major determinant for PCSK9 binding. PCSK9 partly colocalized with hypersulfated heparan sulfate in proteinuric rats, but not in control rats. Hence, proteinuria induces hypersulfated hepatic heparan sulfate proteoglycans, increasing their affinity to PCSK9. This might impair hepatic triglyceride-rich remnant lipoproteins uptake, causing proteinuria-associated dyslipidemia. Thus, our study reveals PCSK9/heparan sulfate may be a novel target to control dyslipidemia.
Collapse
Affiliation(s)
- Pragyi Shrestha
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Saleh Yazdani
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, Leuven, Belgium; Vlaams Institute of Biotechnology Leuven Center for Microbiology, Leuven, Belgium
| | - Romain R Vivès
- University Grenoble Alpes, Institute of Structural Biology (IBS), Atomic Energy and Alternative Energies Commission (CEA), French National Centre for Scientific Research (CNRS), Grenoble, France
| | - Rana El Masri
- University Grenoble Alpes, Institute of Structural Biology (IBS), Atomic Energy and Alternative Energies Commission (CEA), French National Centre for Scientific Research (CNRS), Grenoble, France
| | - Wendy Dam
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jacob van den Born
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
36
|
Shen H, Feng S, Lu Y, Jiang L, Yang T, Wang Z. Correlation between plasma proprotein convertase subtilisin/kexin type 9 and blood lipids in patients with newly diagnosed primary nephrotic syndrome. Ren Fail 2021; 42:405-412. [PMID: 32349585 PMCID: PMC7241483 DOI: 10.1080/0886022x.2020.1756846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a major post-transcriptional regulator of low-density lipoprotein receptor degradation. Recently, PCSK9 was shown to be overexpressed by liver cells in rats with proteinuria. However, the levels of PCSK9 in newly diagnosed primary nephrotic syndrome (PNS) patients and correlations involving PCSK9 and blood lipids are not clearly understood. Methods One hundred and sixteen patients who were newly diagnosed with PNS were enrolled in this study. Results Plasma PCSK9 levels in PNS patients were significantly higher than those in healthy controls [310.86 (250.87, 390.25) ng/ml vs 255.67 (202.26, 320.26) ng/ml, p = 0.002]. Plasma PCSK9 in PNS patients was positively correlated with total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) (γ = 0.246, p = 0.008, and γ = 0.183, p = 0.049). When plasma PCSK9 was >267.60 ng/ml, the risk of developing hypercholesterolemia significantly increased in PNS patients (OR = 6.40, 95% CI 2.06–19.87, p = 0.001). When plasma PCSK9 was >255.05 ng/ml, the risk of developing higher levels of LDL-C significantly increased in PNS patients (OR = 3.83, 95%CI 1.25–11.68, p = 0.018). Conclusions Plasma PCSK9 levels in newly diagnosed PNS patients were markedly increased, and elevated PCSK9 abundance was positively correlated with elevated serum TC and LDL-C levels, suggesting that PCSK9 may emerge as a novel therapeutic target in NS-associated hypercholesterolemia.
Collapse
Affiliation(s)
- Huaying Shen
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng Feng
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Lu
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Linsen Jiang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tingting Yang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi Wang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Zhang PN, Zhou MQ, Guo J, Zheng HJ, Tang J, Zhang C, Liu YN, Liu WJ, Wang YX. Mitochondrial Dysfunction and Diabetic Nephropathy: Nontraditional Therapeutic Opportunities. J Diabetes Res 2021; 2021:1010268. [PMID: 34926696 PMCID: PMC8677373 DOI: 10.1155/2021/1010268] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetic nephropathy (DN) is a progressive microvascular diabetic complication. Growing evidence shows that persistent mitochondrial dysfunction contributes to the progression of renal diseases, including DN, as it alters mitochondrial homeostasis and, in turn, affects normal kidney function. Pharmacological regulation of mitochondrial networking is a promising therapeutic strategy for preventing and restoring renal function in DN. In this review, we have surveyed recent advances in elucidating the mitochondrial networking and signaling pathways in physiological and pathological contexts. Additionally, we have considered the contributions of nontraditional therapy that ameliorate mitochondrial dysfunction and discussed their molecular mechanism, highlighting the potential value of nontraditional therapies, such as herbal medicine and lifestyle interventions, in therapeutic interventions for DN. The generation of new insights using mitochondrial networking will facilitate further investigations on nontraditional therapies for DN.
Collapse
Affiliation(s)
- Ping Na Zhang
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Meng Qi Zhou
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Jing Guo
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Hui Juan Zheng
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Jingyi Tang
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Chao Zhang
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Yu Ning Liu
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| | - Wei Jing Liu
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
- Institute of Nephrology and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, Zhanjiang, China
| | - Yao Xian Wang
- Renal Research Institution of Beijing University of Chinese Medicine and Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Shipping Warehouse No. 5, Beijing 100700, China
| |
Collapse
|
38
|
Mamedov M, Karimov A. Secondary hyperlipidemia: features of manifestation in various somatic diseases. PROFILAKTICHESKAYA MEDITSINA 2021; 24:105. [DOI: 10.17116/profmed202124031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
|
39
|
Busuioc RM, Covic A, Kanbay M, Banach M, Burlacu A, Mircescu G. Protein convertase subtilisin/kexin type 9 biology in nephrotic syndrome: implications for use as therapy. Nephrol Dial Transplant 2020; 35:1663-1674. [PMID: 31157893 DOI: 10.1093/ndt/gfz108] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/19/2019] [Indexed: 01/12/2023] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) levels almost constantly increased in patients with nephrotic syndrome (NS). Protein convertase subtilisin/kexin type 9 (PCSK9) [accelerates LDL-receptor (LDL-R) degradation] is overexpressed by liver cells in NS. Their levels, correlated inversely to LDL-R expression and directly to LDL-C, seem to play a central role in hypercholesterolaemia in NS. Hypersynthesis resulting from sterol regulatory element-binding protein dysfunction, hyperactivity induced by c-inhibitor of apoptosis protein expressed in response to stimulation by tumour necrosis factor-α produced by damaged podocytes and hypo-clearance are the main possible mechanisms. Increased LDL-C may damage all kidney cell populations (podocytes, mesangial and tubular cells) in a similar manner. Intracellular cholesterol accumulation produces oxidative stress, foam cell formation and apoptosis, all favoured by local inflammation. The cumulative effect of cellular lesions is worsened proteinuria and kidney function loss. Accordingly, NS patients should be considered high risk and treated by lowering LDL-C. However, there is still not enough evidence determining whether lipid-lowering agents are helpful in managing dyslipidaemia in NS. Based on good efficacy and safety proved in the general population, therapeutic modulation of PCSK9 via antibody therapy might be a reasonable solution. This article explores the established and forthcoming evidence implicating PCSK9 in LDL-C dysregulation in NS.
Collapse
Affiliation(s)
| | - Adrian Covic
- "Gr. T. Popa," University of Medicine and Pharmacy, Iasi, Romania.,Nephrology Clinic, Dialysis and Renal Transplant Center - 'C.I. Parhon' University Hospital Iasi, Romania
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland.,Polish Mother's Memorial Hospital Research Institute, Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Alexandru Burlacu
- "Gr. T. Popa," University of Medicine and Pharmacy, Iasi, Romania.,Head of Department of Interventional Cardiology - Cardiovascular Diseases Institute Iasi, Romania
| | - Gabriel Mircescu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
40
|
Artunc F. Kidney-derived PCSK9-a new driver of hyperlipidemia in nephrotic syndrome? Kidney Int 2020; 98:1393-1395. [PMID: 33276863 DOI: 10.1016/j.kint.2020.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/27/2022]
Abstract
Increased plasma concentrations of proprotein convertase subtilisin/kexin type 9 or PCSK9, which reduces hepatic uptake of low-density lipoprotein by downregulation of the low-density lipoprotein receptor, have been reported in nephrotic patients and might contribute to hyperlipidemia in nephrotic syndrome. The results of the study by Molina-Jijon et al. found that renal PCSK9 expression was upregulated in the collecting duct of nephrotic patients and animals, suggesting that the kidney might be a major source for plasma PCSK9 in nephrotic syndrome.
Collapse
Affiliation(s)
- Ferruh Artunc
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD) at the University Tübingen, Tübingen, Germany.
| |
Collapse
|
41
|
PCSK9: Associated with cardiac diseases and their risk factors? Arch Biochem Biophys 2020; 704:108717. [PMID: 33307067 DOI: 10.1016/j.abb.2020.108717] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022]
Abstract
PCSK9 plays a critical role in cholesterol metabolism via the PCSK9-LDLR axis. Liver-derived, circulating PCSK9 has become a novel drug target in lipid-lowering therapy. Accumulative evidence supports the possible association between PCSK9 and cardiac diseases and their risk factors. PCSK9 exerts various effects in the heart independently of LDL-cholesterol regulation. Acute myocardial infarction (AMI) induces local and systemic inflammation and reactive oxygen species generation, resulting in increased PCSK9 expression in hepatocytes and cardiomyocytes. PCSK9 upregulation promotes excessive autophagy and apoptosis in cardiomyocytes, thereby contributing to cardiac insufficiency. PCSK9 might also participate in the pathophysiology of heart failure by regulating fatty acid metabolism and cardiomyocyte contractility. It also promotes platelet activation and coagulation in patients with atrial fibrillation. PCSK9 is an independent predictor of aortic valve calcification and accelerates calcific aortic valve disease by regulating lipoprotein(a) catabolism. Accordingly, the use of PCSK9 inhibitors significantly reduced infarct sizes and arrhythmia and improves cardiac contractile function in a rat model of AMI. Circulating PCSK9 levels are positively correlated with age, diabetes mellitus, obesity, and hypertension. Here, we reviewed recent clinical and experimental studies exploring the association between PCSK9, cardiac diseases, and their related risk factors and aiming to identify possible underlying mechanisms.
Collapse
|
42
|
Schlüter KD, Wolf A, Schreckenberg R. Coming Back to Physiology: Extra Hepatic Functions of Proprotein Convertase Subtilisin/Kexin Type 9. Front Physiol 2020; 11:598649. [PMID: 33364976 PMCID: PMC7750466 DOI: 10.3389/fphys.2020.598649] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022] Open
Abstract
Neuronal apoptosis regulated convertase-1 (NARC-1), now mostly known as proprotein convertase subtilisin/kexin type 9 (PCSK9), has received a lot of attention due to the fact that it is a key regulator of the low-density lipoprotein (LDL) receptor (LDL-R) and is therefore involved in hepatic LDL clearance. Within a few years, therapies targeting PCSK9 have reached clinical practice and they offer an additional tool to reduce blood cholesterol concentrations. However, PCSK9 is almost ubiquitously expressed in the body but has less well-understood functions and target proteins in extra hepatic tissues. As such, PCSK9 is involved in the regulation of neuronal survival and protein degradation, it affects the expression of the epithelial sodium channel (ENaC) in the kidney, it interacts with white blood cells and with cells of the vascular wall, and it modifies contractile activity of cardiomyocytes, and contributes to the regulation of cholesterol uptake in the intestine. Moreover, under stress conditions, signals from the kidney and heart can affect hepatic expression and thereby the plasma concentration of PCSK9 which then in turn can affect other target organs. Therefore, there is an intense relationship between the local (autocrine) and systemic (endocrine) effects of PCSK9. Although, PCSK9 has been recognized as a ubiquitously expressed modifier of cellular function and signaling molecules, its physiological role in different organs is not well-understood. The current review summarizes these findings.
Collapse
Affiliation(s)
| | - Annemarie Wolf
- Institute of Physiology, Justus-Liebig-University, Gießen, Germany
| | | |
Collapse
|
43
|
Effective improvement of minimal change nephrotic syndrome with uncontrollable high low-density lipoprotein cholesterol level using evolocumab accompanied by the development of acute pancreatitis. CEN Case Rep 2020; 9:333-337. [DOI: 10.1007/s13730-020-00481-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/17/2020] [Indexed: 10/24/2022] Open
|
44
|
Jatem E, Lima J, Montoro B, Torres-Bondia F, Segarra A. Efficacy and Safety of PCSK9 Inhibitors in Hypercholesterolemia Associated With Refractory Nephrotic Syndrome. Kidney Int Rep 2020; 6:101-109. [PMID: 33426389 PMCID: PMC7783565 DOI: 10.1016/j.ekir.2020.09.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction Treatment of hypercholesterolemia in refractory nephrotic syndrome remains a therapeutic challenge. There is not enough evidence supporting the efficacy of statins, and these drugs can be associated with an increased incidence of adverse effects. Herein we summarize our clinical experience with 12 patients suffering from refractory nephrotic syndrome with associated vascular disease and uncontrolled hypercholesterolemia despite treatment with statins who were treated with proprotein convertase subtilisin kexin 9 (PCSK9) inhibitors. Methods Twelve adult patients with primary nephrotic syndrome refractory to multiple lines of immunosuppressive treatment who suffered from clinical atheromatous vascular disease were treated with PCSK9 inhibitors according to the prescription guidelines for secondary prevention of cardiovascular events. Eight patients with refractory nephrotic syndrome without vascular disease treated with atorvastatin comprised the control group. Results Four weeks after treatment with PCSK9 inhibitors, a statistically significant decrease in total cholesterol and low-density lipoprotein cholesterol (LDL-C) levels was observed without significant changes in serum albumin levels or proteinuria. The mean LDL-C decrease was 36.8% ± 4.9% mmol/L at 4 weeks and remained unchanged throughout the follow-up period. In the control group, there were no significant changes in the levels of total cholesterol or LDL-C during the follow-up period. At the diagnosis of nephrotic syndrome, plasma PCSK9 levels were 334 ± 40 ng/mL and correlated significantly with serum LDL-C levels (r = 0.49, P = 0.023). Six months after starting treatment with PCSK9 inhibitors, plasma PCSK9 levels were significantly reduced to values of 190 ± 36 ng/mL (P = 0.001) with a mean relative reduction of 42.3% ± 12.6%. No local adverse effects were seen at the injection site and no significant changes were seen in the levels of transaminase, creatine phosphokinase, or aldolase. Conclusion PCSK9 inhibitors may be an effective and safe alternative for the treatment of hypercholesterolemia associated with refractory nephrotic syndrome.
Collapse
Affiliation(s)
- Elias Jatem
- Servicio de Nefrología, Hospital Arnau de Vilanova, Lleida, Spain
| | - Joan Lima
- Servicio de Medicina Interna, Hospital Vall d'Hebron, Barcelona, Spain
| | - Bruno Montoro
- Servicio de Farmacia Hospitalaria, Hospital Vall d'Hebron, Barcelona, Spain
| | | | - Alfons Segarra
- Servicio de Nefrología, Hospital Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
45
|
Progress of research on dyslipidemia accompanied by nephrotic syndrome. Chronic Dis Transl Med 2020; 6:182-187. [PMID: 32885154 PMCID: PMC7451721 DOI: 10.1016/j.cdtm.2020.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Indexed: 11/20/2022] Open
Abstract
Nephrotic syndrome is a relatively common clinical disease. Associated dyslipidemia is a risk factor for the occurrence and development of cardiovascular and renal diseases that might gradually develop into atherosclerosis, glomerulosclerosis or tubulointerstitial injury. It also confers an elevated risk of complications such as thromboembolism. If not properly controlled over the long term, dyslipidemia will become a key factor in a poor prognosis. Furthermore, dyslipidemia correlates with an increase in hepatic compensatory synthetic lipoprotein levels and a decrease in lipoprotein clearance, which can be sourced to the downregulation of hepatic and lipoprotein lipase activities in endothelial cells, muscle, and adipose tissue, and clinically characterized as hypertriglyceridemia or hypercholesterolemia. However, further investigations into the mechanism(s) of dyslipidemia are needed, with the resultant detailed perspectives and analyses substantially aiding the further development of treatment guidelines. Currently, statins represent the most popular type of pharmaceutical intervention because they lower hepatic cholesterol production and promote the absorption of low-density lipoprotein-cholesterol from the bloodstream, followed by second-line and other potential therapies to regulate the expression of specific receptors.
Collapse
|
46
|
Quiroga B, Muñoz Ramos P, Álvarez Chiva V. Efectividad y seguridad del uso de inhibidores de PCSK9 en el tratamiento de la dislipidemia en el paciente con insuficiencia renal. Nefrologia 2020; 40:499-505. [DOI: 10.1016/j.nefro.2020.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/19/2020] [Indexed: 11/25/2022] Open
|
47
|
Hari P, Khandelwal P, Smoyer WE. Dyslipidemia and cardiovascular health in childhood nephrotic syndrome. Pediatr Nephrol 2020; 35:1601-1619. [PMID: 31302760 DOI: 10.1007/s00467-019-04301-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/11/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022]
Abstract
Children with steroid-resistant nephrotic syndrome (SRNS) are exposed to multiple cardiovascular risk factors predisposing them to accelerated atherosclerosis. This risk is negligible in steroid-sensitive nephrotic syndrome, but a substantial proportion of children with SRNS progress to chronic kidney disease, exacerbating the already existing cardiovascular risk. While dyslipidemia is an established modifiable risk factor for cardiovascular disease in adults with NS, it is uncertain to what extent analogous risks exist for children. There is increasing evidence of accelerated atherosclerosis in children with persistently high lipid levels, especially in refractory NS. Abnormalities of lipid metabolism in NS include hypertriglyceridemia and hypercholesterolemia due to elevated apolipoprotein B-containing lipoproteins, decreased lipoprotein lipase and hepatic lipase activity, increased hepatic PCSK9 levels, and reduced hepatic uptake of high-density lipoprotein. Existing guidelines for the management of dyslipidemia in children may be adapted to target lower lipid levels in children with NS, but they will most likely require both lifestyle modifications and pharmacological therapy. While there is a lack of data from randomized controlled trials in children with NS demonstrating the benefit of lipid-lowering drugs, therapies including statins, bile acid sequestrants, fibrates, ezetimibe, and LDL apheresis have all been suggested and/or utilized. However, concerns with the use of lipid-lowering drugs in children include unclear side effect profiles and unknown long-term impacts on neurological development and puberty. The recent introduction of anti-PCSK9 monoclonal antibodies and other therapies targeted to the molecular mechanisms of lipid transport disrupted in NS holds promise for the future treatment of dyslipidemia in NS.
Collapse
Affiliation(s)
- Pankaj Hari
- Division of Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Priyanka Khandelwal
- Division of Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - William E Smoyer
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
48
|
Molina-Jijon E, Gambut S, Macé C, Avila-Casado C, Clement LC. Secretion of the epithelial sodium channel chaperone PCSK9 from the cortical collecting duct links sodium retention with hypercholesterolemia in nephrotic syndrome. Kidney Int 2020; 98:1449-1460. [PMID: 32750454 DOI: 10.1016/j.kint.2020.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/08/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
The proprotein PCSK9 functions as a chaperone for the epithelial sodium channel in the cortical collecting duct (CCD), is highly expressed in the liver, and plays a significant role in the pathogenesis of hypercholesterolemia. Lower levels of PCSK9 expression also occur in the normal kidney and intestine. Here, we found increased PCSK9 expression in the CCD of biopsies of patients with primary glomerular disease and explored a possible relationship with hypercholesterolemia of nephrotic syndrome. Significantly elevated serum PCSK9 and cholesterol levels were noted in two models of focal and segmental glomerulosclerosis, the Rrm2b-/- mouse and the Buffalo/Mna rat. Increased expression of PCSK9 in the kidney occurred when liver expression was reduced in both models. The impact of reduced or increased PCSK9 in the CCD on hypercholesterolemia in nephrotic syndrome was next studied. Mice with selective deficiency of PCSK9 expression in the collecting duct failed to develop hypercholesterolemia after injection of nephrotoxic serum. Blocking epithelial sodium channel activity with Amiloride in Rrm2b-/- mice resulted in increased expression of its chaperone PCSK9 in the CCD, followed by elevated plasma levels and worsening hypercholesterolemia. Thus, our data suggest that PCSK9 in the kidney plays a role in the initiation of hypercholesterolemia in nephrotic syndrome and make a case for depletion of PCSK9 early in patients with nephrotic syndrome to prevent the development of hypercholesterolemia.
Collapse
Affiliation(s)
- Eduardo Molina-Jijon
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Stéphanie Gambut
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Camille Macé
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Carmen Avila-Casado
- Department of Pathology, Toronto General Hospital, University of Toronto, Toronto, Canada
| | - Lionel C Clement
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA.
| |
Collapse
|
49
|
Ge M, Fontanesi F, Merscher S, Fornoni A. The Vicious Cycle of Renal Lipotoxicity and Mitochondrial Dysfunction. Front Physiol 2020; 11:732. [PMID: 32733268 PMCID: PMC7358947 DOI: 10.3389/fphys.2020.00732] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
The kidney is one of the most energy-demanding organs that require abundant and healthy mitochondria to maintain proper function. Increasing evidence suggests a strong association between mitochondrial dysfunction and chronic kidney diseases (CKDs). Lipids are not only important sources of energy but also essential components of mitochondrial membrane structures. Dysregulation of mitochondrial oxidative metabolism and increased reactive oxygen species (ROS) production lead to compromised mitochondrial lipid utilization, resulting in lipid accumulation and renal lipotoxicity. However, lipotoxicity can be either the cause or the consequence of mitochondrial dysfunction. Imbalanced lipid metabolism, in turn, can hamper mitochondrial dynamics, contributing to the alteration of mitochondrial lipids and reduction in mitochondrial function. In this review, we summarize the interplay between renal lipotoxicity and mitochondrial dysfunction, with a focus on glomerular diseases.
Collapse
Affiliation(s)
- Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Flavia Fontanesi
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
50
|
Lim CC, Gardner D, Ng RZ, Chin YM, Tan HZ, Mok IY, Choo JC. Synergistic impact of pre-diabetes and immunosuppressants on the risk of diabetes mellitus during treatment of glomerulonephritis and renal vasculitis. Kidney Res Clin Pract 2020; 39:172-179. [PMID: 32541094 PMCID: PMC7321669 DOI: 10.23876/j.krcp.20.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/09/2020] [Accepted: 04/26/2020] [Indexed: 01/27/2023] Open
Abstract
Background Glomerulonephritis is often treated with kidney-saving, but potentially diabetogenic immunosuppressants such as glucocorticosteroids and calcineurin inhibitors. Unfortunately, there are little data on dysglycemia before and after diagnosis and during treatment of glomerulonephritis. We aimed to evaluate the occurrence and risk factors for pre-diabetes and incident diabetes among non-diabetic patients with glomerular disease with or without treatment with immunosuppressants. Methods A single-center, retrospective cohort study was performed on 229 non-diabetic immunosuppressant-naïve adults diagnosed with glomerulonephritis and renal vasculitis. Patients with known diabetes and prior immunosuppressant treatment were excluded. Outcomes of new-onset pre-diabetes and new-onset diabetes were defined according to American Diabetic Association criteria. Results Pre-diabetes was present pre-biopsy in 74 of the 229 patients (32.3%). During the median follow-up of 34.0 (23.3-47.5) months, 29 patients (12.7%) developed new-onset diabetes and 58 (25.3%) had new-onset pre-diabetes. Immunosuppressive therapy in patients with pre-existing pre-diabetes was associated with increased odds of new-onset diabetes compared to those without either risk factor (26.0% versus 5.0%; odds ratio, 6.67; 95% confidence interval [CI], 1.41 to 31.64), P = 0.02). Conclusion New-onset diabetes after immunosuppressant treatment occurred in one-quarter of patients with glomerulonephritis and pre-existing pre-diabetes. Physicians should screen for pre-diabetes when planning treatment with immunosuppressants, as its presence significantly increases the risk of diabetes mellitus.
Collapse
Affiliation(s)
| | - Daphne Gardner
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Rui Zhi Ng
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Yok Mooi Chin
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Hui Zhuan Tan
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Irene Yj Mok
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Jason Cj Choo
- Department of Renal Medicine, Singapore General Hospital, Singapore
| |
Collapse
|