1
|
Sun Y, Li Z, Feng N. Association of non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and chronic kidney disease in elderly insights from NHANES. Sci Rep 2025; 15:12611. [PMID: 40221576 PMCID: PMC11993742 DOI: 10.1038/s41598-025-96299-7] [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: 10/22/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) is a novel lipid index. Prior research has established a connection between lipid irregularities and chronic kidney disease (CKD). This study aims to establish a possible link between NHHR and CKD. Data from the National Health and Nutrition Examination Survey (NHANES) spanning from 2003 to 2016 was used to examine the relationship between NHHR and CKD among the elderly population. This research utilized weighted logistic regression, smoothed curve fitting and subgroup analyses along with interaction tests to evaluate the association between NHHR and CKD. The findings reveal a positive correlation between NHHR and CKD in fully adjusted Model 3. Besides, NHHR had a J-curve relationship with CKD. Subgroup analysis indicated that compared with those with lower body mass index (BMI), individuals with higher BMI are more prone to CKD. Research has shown that increased NHHR levels are associated with a higher likelihood of developing CKD in individuals aged above 60 in the United States. NHHR's role in lipid metabolism suggests it might be an effective marker for tracking CKD's progression.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Zhou Li
- Department of Urology, Nanjing Medical University, Wuxi No.2 Hospital, Wuxi, China
| | - Ninghan Feng
- Department of Urology, Jiangnan University Medical Center, Wuxi, China.
- Department of Urology, Nanjing Medical University, Wuxi No.2 Hospital, Wuxi, China.
- Jiangnan University Medical Center, 68 Zhongshan Road, Wuxi, Jiangsu, China.
| |
Collapse
|
2
|
Kosugi T, Eriguchi M, Yoshida H, Tamaki H, Uemura T, Tasaki H, Furuyama R, Fukata F, Nishimoto M, Matsui M, Samejima KI, Iseki K, Fujimoto S, Konta T, Moriyama T, Yamagata K, Narita I, Kasahara M, Shibagaki Y, Kondo M, Asahi K, Watanabe T, Tsuruya K. Serum High-Density Lipoprotein Cholesterol Levels and the Risk of Kidney Function Decline: The Japan Specific Health Checkups (J‑SHC) Study. J Atheroscler Thromb 2025; 32:407-420. [PMID: 39313383 PMCID: PMC11973525 DOI: 10.5551/jat.65107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024] Open
Abstract
AIMS Both low and high serum levels of high-density lipoprotein cholesterol (HDL-C) were reported to be associated with adverse kidney outcomes. However, this association has not been well investigated in the general Japanese population. METHODS This nationwide longitudinal study used data from the Japan Specific Health Checkups Study conducted between 2008-2014. The association between serum HDL-C levels and 40% decline in estimated glomerular filtration rate (eGFR) was analyzed using Cox regression analysis. Trajectories of eGFR were compared using mixed-effects model. RESULTS Among 768,495 participants, 6,249 developed 40% decline in eGFR during the median follow-up period of 34.6 (interquartile range: 14.8-48.4) months. Using serum HDL-C levels of 40-59 mg/dL as a reference, the adjusted hazard ratios (95% confidence intervals) for the kidney outcome of serum HDL-C levels of <40, 60-79 and ≥ 80 mg/dL were 1.26 (1.14-1.39), 0.91 (0.86-0.96), and 0.86 (0.78-0.93), respectively. Restricted cubic spline analysis showed that HDL-C levels of less than approximately 60 mg/dL were associated with an increased risk of kidney outcomes. Subgroup analysis showed that baseline eGFR and proteinuria modified the effects of serum HDL-C levels on kidney outcomes. The mixed-effects model showed that the lower category of HDL-C level was associated with a higher eGFR decline rate (p for interaction <0.001). CONCLUSIONS Low HDL-C levels were associated with kidney function decline; however, high HDL-C levels were not associated with adverse kidney outcomes in the general Japanese population.
Collapse
Affiliation(s)
- Takaaki Kosugi
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Masahiro Eriguchi
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Osaka, Japan
| | - Hiroyuki Tamaki
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Takayuki Uemura
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Hikari Tasaki
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Riri Furuyama
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Fumihiro Fukata
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | | | - Masaru Matsui
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Ken-ichi Samejima
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Kunitoshi Iseki
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Shouichi Fujimoto
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Tsuneo Konta
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Toshiki Moriyama
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Kunihiro Yamagata
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Ichiei Narita
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Masato Kasahara
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Yugo Shibagaki
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Masahide Kondo
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Koichi Asahi
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Tsuyoshi Watanabe
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| |
Collapse
|
3
|
Li Q, Lan W. L-shaped association between dietary niacin intake and chronic kidney disease among adults in the USA: a cross-sectional study. Ren Fail 2024; 46:2399742. [PMID: 39238253 PMCID: PMC11382734 DOI: 10.1080/0886022x.2024.2399742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/03/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD), which has become a global public health issue, is associated with mitochondrial dysfunction. Niacin is a necessary coenzyme for mitochondrial energy metabolism. However, the association between dietary niacin intake and CKD remains uncertain. This study aimed to investigate the association between dietary niacin intake and CKD in American adults. METHODS This is a cross-sectional study. 25,608 individuals aged ≥20 years from the National Health and Nutrition Examination Survey from 2007 to 2018 were involved.Dietary niacin intake was estimated based on 24-hour dietary recalls conducted by trained personnel. CKD was determined by an estimated glomerular filtration rate (eGFR) (<60 ml/min/1.73 m2) or a urinary albumin-to-creatinine ratio (ACR) (≥30mg/g). The association between dietary niacin intake and CKD was investigated using multivariable logistic regression analysis. RESULTS Of 25,608 participants, 17.14% (4388/25,608) had CKD. Compared to individuals with lower niacin intake (quartile [Q]1, ≤15.30 mg/day), those with higher niacin intake in Q2 (15.31-22.07 mg/day), Q3 (22.08-31.09 mg/day), and Q4 (≥31.10 mg/day) exhibited adjusted odds ratios for CKD of 0.89 (95% confidence interval [CI]:0.81-0.99, p = 0.024), 0.83 (95% CI:0.75-0 .92, p < 0 .001), and 0.83 (95% CI:0.75-0.93, p = 0.001) respectively. The relationship between dietary niacin intake and CKD among U.S. adults follows an L-shaped pattern, with an inflection point at approximately 28.04 mg/day. CONCLUSIONS These results suggest an L-shaped association between dietary niacin intake and CKD. Individuals with low dietary niacin intake levels should be alert to the risk of CKD.
Collapse
Affiliation(s)
- Qishu Li
- Department of Nephrology, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| | - Wei Lan
- Department of Nephrology, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Ao Y, Ye H, Liu X, Li Y, Liu H, Ye S, Hu Y, Zhuang P, Zhang Y, Zheng C, Jiao J. Fish oil supplementation in relation to the risk of chronic kidney disease among patients with diabetes. Diabetes Obes Metab 2024; 26:5283-5292. [PMID: 39192528 DOI: 10.1111/dom.15880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024]
Abstract
AIM To investigate the association between fish oil supplementation and subsequent risk of chronic kidney disease (CKD) among patients with diabetes, and further evaluate the mediation effect of typical glycolipid and inflammatory biomarkers. METHODS In total, 24 497 patients with diabetes from the UK Biobank were included. Cox proportional hazards regression models were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for CKD risk, and the rate advancement period was calculated to quantify and communicate the impact of fish oil upon that risk. In addition, we also used mediation analysis to assess the mediating role of plasma biomarkers. RESULTS Overall, 7122 patients reported taking fish oil supplements. During a mean of 11.3 years of follow-up, 3533 CKD cases occurred. In the fully adjusted model, fish oil use was inversely associated with the incidence of CKD (HR 0.90; 95% CI: 0.83, 0.97), which was mediated by serum levels of HbA1c (4.7%), C-reactive protein (CRP) (3.4%) and high-density lipoprotein cholesterol (HDL-C) (2.3%). Participants who took fish oil supplements displayed the same risk of CKD events, but that risk was delayed by approximately 2.79 years compared with non-users of fish oil. CONCLUSIONS Our findings advocate the beneficial role of fish oil use in preventing CKD among patients with diabetes, which may be mediated by serum levels of HbA1c, CRP and HDL-C, and support public health policies aiming to promote fish oil supplementation for the prevention of diabetes complications.
Collapse
Affiliation(s)
- Yang Ao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Ye
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin Li
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyin Liu
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Ye
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yepeng Hu
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory for Agro-Food Processing, Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Chao Zheng
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Al Zein M, Khazzeka A, El Khoury A, Al Zein J, Zoghaib D, Eid AH. Revisiting high-density lipoprotein cholesterol in cardiovascular disease: Is too much of a good thing always a good thing? Prog Cardiovasc Dis 2024; 87:50-59. [PMID: 39442601 DOI: 10.1016/j.pcad.2024.10.009] [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] [Received: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular disease (CVD) continues to be a leading cause of global mortality and morbidity. Various established risk factors are linked to CVD, and modifying these risk factors is fundamental in CVD management. Clinical studies underscore the association between dyslipidemia and CVD, and therapeutic interventions that target low-density lipoprotein cholesterol elicit clear benefits. Despite the correlation between low high-density lipoprotein cholesterol (HDLC) and heightened CVD risk, HDL-raising therapies have yet to showcase significant clinical benefits. Furthermore, evidence from epidemiological and genetic studies reveals that not only low HDL-C levels, but also very high levels of HDL-C are linked to increased risk of CVD. In this review, we focus on HDL metabolism and delve into the relationship between HDL and CVD, exploring HDL functions and the observed alterations in its roles in disease. Altogether, the results discussed herein support the conventional wisdom that "too much of a good thing is not always a good thing". Thus, our recommendation is that a careful reconsideration of the impact of high HDL-C levels is warranted, and shall be revisited in future research.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Alicia Khazzeka
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Dima Zoghaib
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
6
|
Desiana D, Muchlisin ZA, Suhud K, Gani BA. Tribal differences in hypertension and cholesterol profiles in Aceh, Indonesia. Glob Cardiol Sci Pract 2024; 2024:e202422. [PMID: 38983750 PMCID: PMC11230111 DOI: 10.21542/gcsp.2024.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/30/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND One of the factors that contributes to coronary heart disease and stroke is high blood pressure, or hypertension. Hypertension is influenced by race and sex. The objective of this study was to assess the hypertensive population in Aceh by tribal community and to examine the relationship between cholesterol history and hypertension. METHODS This study used incidental sampling as a non-probability sampling method, in which 152 participants were evaluated for the profile of hypertension with a history of cholesterol. Blood pressure was measured using a blood pressure measuring device. HDL, LDL, triglyceride, and total cholesterol levels were measured using LIPID Pro. Data analysis was performed using the Kruskal-Wallis and Mann-Whitney tests with p < 0.05. RESULTS The study population (N = 152) consisted of 81 males (53%) and 71 females (47%) across the ethnicities of Aceh (64:42%), Gayo (19:13%), Alas (33:22%), and Aneuk Jamee (36:24%). In the male group, hypertension was associated with total cholesterol (r = 0.03; p = 0.78), HDL (r = 0.20; p = 0.07), and LDL (r = 0.21; p = 0.07) levels, whereas in the female group, hypertension was primarily correlated with LDL levels (r = 0.20; p = 0.09). CONCLUSION In general, hypertension in males and females in the four tribes in Aceh is associated with HDL, LDL, and total cholesterol levels.
Collapse
Affiliation(s)
- Desiana Desiana
- Graduate School of Faculty of Mathematics and Applied Sciences, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | | | - Khairi Suhud
- Department Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Basri A Gani
- Department of Oral Biology, Dentistry faculty, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| |
Collapse
|
7
|
Lonardo A. Association of NAFLD/NASH, and MAFLD/MASLD with chronic kidney disease: an updated narrative review. METABOLISM AND TARGET ORGAN DAMAGE 2024; 4. [DOI: 10.20517/mtod.2024.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Chronic kidney disease (CKD) and nonalcoholic fatty liver disease (NAFLD), metabolic dysfunction-associated fatty liver disease (MAFLD) and metabolic dysfunction-associated steatotic liver disease (MASLD) account for substantial financial burden worldwide. These alarming features call for enhanced efforts to prevent and manage the development and progression of CKD. Accumulating evidence supporting a causal role of NAFLD/MAFLD/MASLD-in CKD opens new horizons to achieve this aim. Recent epidemiological studies and meta-analyses exploring the association of NAFLD/MAFLD/MASLD with CKD and the characteristics of NAFLD/MAFLD/MASLD associated with the odds of incident CKD are discussed. The involved pathomechanisms, including the common soil hypothesis, genetics, gut dysbiosis, and portal hypertension, are examined in detail. Finally, lifestyle changes (diet and physical exercise), direct manipulation of gut microbiota, and drug approaches involving statins, renin-angiotensin-aldosterone system inhibitors, GLP-1 Receptor Agonists, Sodium-glucose cotransporter-2, pemafibrate, and vonafexor are examined within the context of prevention and management of CKD among those with NAFLD/MAFLD/MASLD. The evolving NAFLD/MAFLD/MASLD nomenclature may generate confusion among practicing clinicians and investigators. However, comparative studies investigating the pros and contra of different nomenclatures may identify the most useful definitions among NAFLD/MAFLD/MASLD and strategies to identify, prevent, and halt the onset and progression of CKD.
Collapse
|
8
|
Liu P, Li J, Yang L, Zhang Z, Zhao H, Zhao N, Ou W, Zhang Y, Chen S, Wang G, Zhang X, Wu S, Yang X. Association between cumulative uric acid to high-density lipoprotein cholesterol ratio and the incidence and progression of chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1269580. [PMID: 38155948 PMCID: PMC10753577 DOI: 10.3389/fendo.2023.1269580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Objective The ratio of uric acid to high-density lipoprotein cholesterol (UHR) was related to the risk of chronic kidney disease (CKD), we aimed to investigate the association of cumulative UHR (cumUHR) with incidence and progression of CKD. Methods Our study included a total of 49,913 participants (mean age 52.57 years, 77% males) from the Kailuan Study conducted between 2006 and 2018. Participants who completed three consecutive physical examinations were included. Cumulative UHR (cumUHR) was computed as the summed average UHR between two consecutive physical examinations, multiplied by the time between the two examinations. Participants were then categorized into four groups based on cumUHR quartiles. Subsequently, participants were further divided into a CKD group and a non-CKD group. The associations between cumUHR and CKD and it's progression were assessed by Cox proportional hazards regression models. The cumulative incidence of endpoint events was compared between the cumUHR groups using the log-rank test. The C-index, net reclassification improvement (NRI) and integrated discrimination improvement (IDI) were calculated to assess the predictive performance of cumUHR. Results After a mean follow-up of 8.0 ± 1.7 years, there were 4843 cases of new-onset CKD, 2504 of low eGFR, and 2617 of proteinuria in the non-CKD group. Within the CKD group, there were 1952 cases of decline in eGFR category, 1465 of >30% decline in eGFR, and 2100 of increased proteinuria. In the non-CKD group, the adjusted hazard ratios (HRs) and confidence intervals (CIs) in the fourth quartile were 1.484 (1.362-1.617), 1.643 (1.457-1.852), and 1.324 (1.179-1.486) for new-onset CKD, low eGFR, and proteinuria, respectively. In the CKD group, the adjusted HRs in the fourth quartile were 1.337 (1.164-1.534), 1.428 (1.216-1.677), and 1.446 (1.267-1.651) for decline in eGFR category, >30% decline in eGFR, and increase in proteinuria, respectively. In addition, we separately added a single UHR measurement and cumUHR to the CKD base prediction model and the CKD progression base prediction model, and found that the models added cumUHR had the highest predictive value. Conclusion High cumUHR exposure was an independent risk factor for the incidence and progression of CKD, and it was a better predictor than a single UHR measurement.
Collapse
Affiliation(s)
- Peipei Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Junjuan Li
- Department of Nephrology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Ling Yang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Zihao Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Hua Zhao
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
| | - Naihui Zhao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wenli Ou
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yinggen Zhang
- Department of Nuclear Medicine, Kailuan General Hospital, Tangshan, Hebei, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Guodong Wang
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Xiaofu Zhang
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Xiuhong Yang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
9
|
Vitali C, Pavanello C, Turri M, Lund-Katz S, Phillips MC, Catapano AL, Baragetti A, Norata GD, Veglia F, Calabresi L. Apolipoprotein E isoforms differentially affect LCAT-dependent cholesterol esterification. Atherosclerosis 2023; 382:117266. [PMID: 37725860 DOI: 10.1016/j.atherosclerosis.2023.117266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND AND AIMS LCAT esterifies cholesterol in both HDL (α-activity) and apoB-containing lipoproteins (β-activity). The main activator of LCAT β-activity is apoE, which in humans exists in 3 main different isoforms (E2, E3 and E4). Here, to gather insights into the potential role of LCAT in apoB-containing lipoprotein metabolism, we investigated the ability of apoE isoforms to promote LCAT-mediated cholesterol esterification. METHODS We evaluated the plasma cholesterol esterification rate (CER) in 311 individuals who express functional LCAT and either apoE2, apoE3, or apoE4 and in 28 individuals who also carried LCAT mutations causing selective loss of LCAT α-activity (Fish-Eye Disease (FED)-causing mutations). The association of carrier status with CER was determined using an adjusted linear regression model. The kinetic of LCAT activity towards reconstituted HDLs (rHDLs) containing each apoE isoform was determined using the Michaelis-Menten model. RESULTS Plasma CER was ∼20% higher in apoE2 carriers compared to apoE3 carriers, and ∼30% higher in apoE2 carriers compared to apoE4 carriers. After adjusting for age, sex, total cholesterol, HDL-C, apoA-I, apoB, chronic kidney disease diagnosis, zygosity, and LCAT concentration, CER remained significantly different among carriers of the three apoE isoforms. The same trend was observed in carriers of FED-causing mutations. rHDLs containing apoE2 were associated with a lower affinity but higher maximal esterification rate, compared to particles containing apoE3 or apoE4. CONCLUSION The present results suggest that the apoE2 isoform is associated with a higher LCAT-mediated cholesterol esterification. This observation may contribute to the characterization of the peculiar functional properties of apoE2.
Collapse
Affiliation(s)
- Cecilia Vitali
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Marta Turri
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Sissel Lund-Katz
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Phillips
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Andrea Baragetti
- IRCCS Multimedica, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
10
|
van Zonneveld AJ, Zhao Q, Rotmans JI, Bijkerk R. Circulating non-coding RNAs in chronic kidney disease and its complications. Nat Rev Nephrol 2023; 19:573-586. [PMID: 37286733 DOI: 10.1038/s41581-023-00725-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/09/2023]
Abstract
Post-transcriptional regulation by non-coding RNAs (ncRNAs) can modulate the expression of genes involved in kidney physiology and disease. A large variety of ncRNA species exist, including microRNAs, long non-coding RNAs, piwi-interacting RNAs, small nucleolar RNAs, circular RNAs and yRNAs. Despite early assumptions that some of these species may exist as by-products of cell or tissue injury, a growing body of literature suggests that these ncRNAs are functional and participate in a variety of processes. Although they function intracellularly, ncRNAs are also present in the circulation, where they are carried by extracellular vesicles, ribonucleoprotein complexes or lipoprotein complexes such as HDL. These systemic, circulating ncRNAs are derived from specific cell types and can be directly transferred to a variety of cells, including endothelial cells of the vasculature and virtually any cell type in the kidney, thereby affecting the function of the host cell and/or its response to injury. Moreover, chronic kidney disease itself, as well as injury states associated with transplantation and allograft dysfunction, is associated with a shift in the distribution of circulating ncRNAs. These findings may provide opportunities for the identification of biomarkers with which to monitor disease progression and/or the development of therapeutic interventions.
Collapse
Affiliation(s)
- Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Qiao Zhao
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands.
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
11
|
Pavanello C, Ossoli A. HDL and chronic kidney disease. ATHEROSCLEROSIS PLUS 2023; 52:9-17. [PMID: 37193017 PMCID: PMC10182177 DOI: 10.1016/j.athplu.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023]
Abstract
Low HDL-cholesterol (HDL-C) concentrations are a typical trait of the dyslipidemia associated with chronic kidney disease (CKD). In this condition, plasma HDLs are characterized by alterations in structure and function, and these particles can lose their atheroprotective functions, e.g., the ability to promote cholesterol efflux from peripheral cells, anti-oxidant and anti-inflammatory proprieties and they can even become dysfunctional, i.e., exactly damaging. The reduction in plasma HDL-C levels appears to be the only lipid alteration clearly linked to the progression of renal disease in CKD patients. The association between the HDL system and CKD development and progression is also supported by the presence of genetic kidney alterations linked to HDL metabolism, including mutations in the APOA1, APOE, APOL and LCAT genes. Among these, renal disease associated with LCAT deficiency is well characterized and lipid abnormalities detected in LCAT deficiency carriers mirror the ones observed in CKD patients, being present also in acquired LCAT deficiency. This review summarizes the major alterations in HDL structure and function in CKD and how genetic alterations in HDL metabolism can be linked to kidney dysfunction. Finally, the possibility of targeting the HDL system as possible strategy to slow CKD progression is reviewed.
Collapse
Affiliation(s)
| | - Alice Ossoli
- Corresponding author. Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133, Milano, Italy.
| |
Collapse
|
12
|
Pazarín-Villaseñor L, García-Salas Y, Yanowsky-Escatell FG, Pacheco-Moisés FP, Andrade-Sierra J, Campos-Bayardo TI, Román-Rojas D, García-Sánchez A, Miranda-Díaz AG. Oxidation State in Peritoneal Dialysis in Patients with Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24032669. [PMID: 36768992 PMCID: PMC9916940 DOI: 10.3390/ijms24032669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
End-stage renal disease (ESRD) progression is closely related to oxidative stress (OS). The study objective was to determine the oxidant and antioxidant status in peritoneal dialysis (PD) patients with type 2 diabetes mellitus (DM). An analytical cross-sectional study from the PD program was carried out with 62 patients, 22 with and 40 without DM. Lipoperoxides (LPO) levels in patients with DM, 3.74 ± 1.09 mM/L, and without DM, 3.87 ± 0.84 mM/L were found to increase compared to healthy controls (HC) 3.05 ± 0.58 mM/L (p = 0.006). The levels of the oxidative DNA damage marker (8-OH-dG) were found to be significantly increased in patients with DM, 1.71 ng/mL (0.19-71.92) and without DM, 1.05 ng/mL (0.16-68.80) front to 0.15 ng/mL (0.15-0.1624) of HC (p = 0.001). The antioxidant enzyme superoxide dismutase (SOD) activity was found to be significantly increased in patients with DM, 0.37 ± 0.15 U/mL, and without DM, 0.37 ± 0.17 compared to HC, 0.23 ± 0.05 U/mL (p = 0.038). The activity of the enzyme glutathione peroxidase (GPx) showed a significant increase (p < 0.001) in patients with DM, 3.56 ± 2.18 nmol/min/mL, and without DM, 3.28 ± 1.46 nmol/min/mL, contrary to the activity obtained in HC, 1.55 ± 0.34 nmol/min/mL. In conclusion, we found an imbalance of oxidative status in patients undergoing PD with and without DM through the significant increase in LPO oxidants and the marker of oxidative damage in DNA. The activity of the antioxidant enzymes SOD and GPx were significantly increased in patients with and without DM undergoing PD, possibly in an attempt to compensate for the deregulation of oxidants. Antioxidant enzymes could be promising therapeutic strategies as a complement to the management of chronic kidney diseases.
Collapse
Affiliation(s)
- Leonardo Pazarín-Villaseñor
- Nephrology Service, Civil Hospital of Guadalajara “Dr. Juan I Menchaca”, Guadalajara 44280, Jalisco, Mexico
- Nephrology Specialty, Regional General Hospital No. 46 of the IMSS, Guadalajara 44910, Jalisco, Mexico
| | - Yessica García-Salas
- Nephrology Service, Civil Hospital of Guadalajara “Dr. Juan I Menchaca”, Guadalajara 44280, Jalisco, Mexico
| | - Francisco Gerardo Yanowsky-Escatell
- Nephrology Service, Civil Hospital of Guadalajara “Dr. Juan I Menchaca”, Guadalajara 44280, Jalisco, Mexico
- Department of Health Sciences-Illness as an Individual Process, University Center of Tonala, University of Guadalajara, Guadalajara 45425, Jalisco, Mexico
| | - Fermín Paul Pacheco-Moisés
- Department of Chemistry, University Centre for Exact and Engineering Sciences, University of Guadalajara, Guadalajara 44430, Jalisco, Mexico
| | - Jorge Andrade-Sierra
- Nephrology Service, Civil Hospital of Guadalajara “Dr. Juan I Menchaca”, Guadalajara 44280, Jalisco, Mexico
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico
| | - Tannia Isabel Campos-Bayardo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico
| | - Daniel Román-Rojas
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico
| | - Andrés García-Sánchez
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
13
|
Magnoni M, Andreini D, Pirillo A, Uboldi P, Latini R, Catapano AL, Maggioni AP, Norata GD. Predictive value of HDL function in patients with coronary artery disease: relationship with coronary plaque characteristics and clinical events. Ann Med 2022; 54:1036-1046. [PMID: 35438019 PMCID: PMC9090377 DOI: 10.1080/07853890.2022.2063374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND HDL is endowed with several metabolic, vascular, and immunoinflammatory protective functions. Among them, a key property is to promote reverse cholesterol transport from cells back to the liver. The aim of this study was to estimate the association of scavenger receptor class B type I (SR-BI)- and ATP binding cassette transporter A1 (ABCA1)-mediated cholesterol efflux (the two major routes for cholesterol efflux to HDL) with the presence, extent, and severity of coronary artery disease (CAD), vascular wall remodelling processes, coronary plaque characteristics, and the incidence of myocardial infarction in the different subgroups of patients from the CAPIRE study. METHODS Patients (n = 525) from the CAPIRE study were divided into two groups: low-risk factors (RF), with 0-1 RF (n = 263), and multiple-RF, with ≥2 RFs; within each group, subjects were classified as no-CAD or CAD based on the segment involvement score (SIS) evaluated by coronary computed tomography angiography (SIS = 0 and SIS > 5, respectively). SR-BI- and ABCA1-mediated cholesterol efflux were measured using the plasma of all patients. RESULTS SR-BI-mediated cholesterol efflux was significantly reduced in patients with CAD in both the low-RF and multiple-RF groups, whereas ABCA1-mediated cholesterol efflux was similar among all groups. In CAD patients, multivariable analysis showed that SR-BI-mediated cholesterol efflux <25th percentile predicted cardiovascular outcome (odds ratio 4.1; 95% CI: 1.3-13.7; p = .019), whereas ABCA-1-mediated cholesterol efflux and HDL-C levels significantly did not. Despite this finding, reduced SR-BI-mediated cholesterol efflux was not associated with changes in high-risk plaque features or changes in the prevalence of elevated total, non-calcified, and low-attenuation plaque volume. CONCLUSION SR-BI-mediated cholesterol efflux capacity is lower in patients with diffuse coronary atherosclerosis. In addition, a lower SR-BI-mediated cholesterol efflux capacity is associated with the worst clinical outcomes in patients with CAD, independently of atherosclerotic plaque features. Key MessagesIncreased cholesterol efflux capacity, an estimate of HDL function, is associated with a reduced CVD risk, regardless of HDL-C levels.HDL-C levels are significantly lower in patients with CAD.Lower SR-BI-mediated cholesterol efflux capacity is observed in patients with diffuse coronary atherosclerosis and is associated with the worst clinical outcomes in patients with CAD, independently of atherosclerotic plaque features.
Collapse
Affiliation(s)
| | - Daniele Andreini
- IRCCS, Centro Cardiologico Monzino, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Angela Pirillo
- Centro SISA per lo Studio dell'Aterosclerosi, Ospedale Bassini, Balsamo, Italy.,IRCSS Multimedica, Milan, Italy
| | - Patrizia Uboldi
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Roberto Latini
- Department of Cardiovascular Medicine, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Alberico L Catapano
- IRCSS Multimedica, Milan, Italy.,Department of Excellence of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Aldo P Maggioni
- Heart Care Foundation ANMCO Research Center, Florence, Italy
| | - Giuseppe D Norata
- Centro SISA per lo Studio dell'Aterosclerosi, Ospedale Bassini, Balsamo, Italy.,Department of Excellence of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
14
|
Recent Advances in Understanding of Cardiovascular Diseases in Patients with Chronic Kidney Disease. J Clin Med 2022; 11:jcm11164653. [PMID: 36012887 PMCID: PMC9409994 DOI: 10.3390/jcm11164653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
|
15
|
Li M, Feng S, Zhan X, Peng F, Feng X, Zhou Q, Wu X, Wang X, Su N, Tang X, Wang Z, Zhang Y, Zeng Y, Zhu L, Xie Y, Liang J, Liu L, Wen Y. Neutrophil to high-density lipoprotein ratio associates with higher all-cause mortality and new onset cardiovascular events in peritoneal dialysis patients. Int Urol Nephrol 2022; 54:2745-2754. [PMID: 35411415 DOI: 10.1007/s11255-022-03202-8] [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: 08/21/2021] [Accepted: 03/15/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND Neutrophil to high-density lipoprotein ratio (NHR), a new inflammatory marker, is associated with poor clinical prognosis. However, the correlation of NHR and adverse outcomes in peritoneal dialysis (PD) patients remains unclear. METHODS In this retrospective cohort study, a total of 1051 PD patients were recruited from three centers during Jan 1, 2009 to Dec 31, 2017. Eligible patients were distributed according to quartiles of the NHR. Kaplan-Meier cumulative incidence curves, multivariate COX regression, competitive risk analysis and restricted cubic spline (RCS) were applied to analyze the relationship between NHR and all-cause mortality as well as cardiovascular events (CVE). In addition, forest plots were used to calculate the interaction between different subgroups. RESULTS During follow-up, a total of 240 all-cause mortality and 157 new-onset CVE were recorded. The all-cause mortality in the highest quartile of NHR (> 5.43) were higher than those in the other groups. RCS showed a non-linear relationship between NHR and adverse outcomes. Multivariate COX regression indicated elevated NHR was an independent risk factor for all-cause mortality. Compared to the highest quartile, hazard ratio (HR) of new-onset CVE equals to 0.522 (95% CI 0.321-0.849) in the secondary quartile (2.43 < NHR ≤ 3.57), and the HR of all-cause mortality analysis is 0.551 (95% CI 0.378-0.803) in the third quartile (3.57 < NHR ≤ 5.43). Kaplan-Meier analysis suggested there were significant differences in all-cause mortality and new-onset CVE among four NHR groups. CONCLUSIONS NHR was a new independent risk factor for all-cause mortality in PD patients.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaozhen Feng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaojiang Zhan
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoran Feng
- Department of Nephrology, Jiujiang NO.1 people's Hospital, Jiangxi, China
| | - Qian Zhou
- Department of Medical Statistics, Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xianfeng Wu
- Department of Nephrology, Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyang Wang
- Department of Nephrology, The First Affiliated Hospital, Zhengzhou University, ZhengZhou, Henan, China
| | - Ning Su
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xingming Tang
- Department of Nephrology, Affiliated Tungwah Hospital, Sun Yet-Sen University, Dongguan, Guangdong, China
| | - Zebin Wang
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yujing Zhang
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yingsi Zeng
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liya Zhu
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuxin Xie
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianbo Liang
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lingling Liu
- Department of General Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yueqiang Wen
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
16
|
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: 2.7] [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]
|
17
|
HDL in Atherosclerotic Cardiovascular Disease: In Search of a Role. Cells 2021; 10:cells10081869. [PMID: 34440638 PMCID: PMC8394469 DOI: 10.3390/cells10081869] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
For a long time, high-density lipoprotein cholesterol (HDL-C) has been regarded as a cardiovascular disease (CVD) protective factor. Recently, several epidemiological studies, while confirming low plasma levels of HDL-C as an established predictive biomarker for atherosclerotic CVD, indicated that not only people at the lowest levels but also those with high HDL-C levels are at increased risk of cardiovascular (CV) mortality. This “U-shaped” association has further fueled the discussion on the pathophysiological role of HDL in CVD. In fact, genetic studies, Mendelian randomization approaches, and clinical trials have challenged the notion of HDL-C levels being causally linked to CVD protection, independent of the cholesterol content in low-density lipoproteins (LDL-C). These findings have prompted a reconsideration of the biological functions of HDL that can be summarized with the word “HDL functionality”, a term that embraces the many reported biological activities beyond the so-called reverse cholesterol transport, to explain this lack of correlation between HDL levels and CVD. All these aspects are summarized and critically discussed in this review, in an attempt to provide a background scenario for the “HDL story”, a lipoprotein still in search of a role.
Collapse
|
18
|
Mohammad S, Al Zoubi S, Collotta D, Krieg N, Wissuwa B, Ferreira Alves G, Purvis GSD, Norata GD, Baragetti A, Catapano AL, Solito E, Zechendorf E, Schürholz T, Correa-Vargas W, Brandenburg K, Coldewey SM, Collino M, Yaqoob MM, Martin L, Thiemermann C. A Synthetic Peptide Designed to Neutralize Lipopolysaccharides Attenuates Metaflammation and Diet-Induced Metabolic Derangements in Mice. Front Immunol 2021; 12:701275. [PMID: 34349763 PMCID: PMC8328475 DOI: 10.3389/fimmu.2021.701275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Metabolic endotoxemia has been suggested to play a role in the pathophysiology of metaflammation, insulin-resistance and ultimately type-2 diabetes mellitus (T2DM). The role of endogenous antimicrobial peptides (AMPs), such as the cathelicidin LL-37, in T2DM is unknown. We report here for the first time that patients with T2DM compared to healthy volunteers have elevated plasma levels of LL-37. In a reverse-translational approach, we have investigated the effects of the AMP, peptide 19-2.5, in a murine model of high-fat diet (HFD)-induced insulin-resistance, steatohepatitis and T2DM. HFD-fed mice for 12 weeks caused obesity, an impairment in glycemic regulations, hypercholesterolemia, microalbuminuria and steatohepatitis, all of which were attenuated by Peptide 19-2.5. The liver steatosis caused by feeding mice a HFD resulted in the activation of nuclear factor kappa light chain enhancer of activated B cells (NF-ĸB) (phosphorylation of inhibitor of kappa beta kinase (IKK)α/β, IκBα, translocation of p65 to the nucleus), expression of NF-ĸB-dependent protein inducible nitric oxide synthase (iNOS) and activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, all of which were reduced by Peptide 19-2.5. Feeding mice, a HFD also resulted in an enhanced expression of the lipid scavenger receptor cluster of differentiation 36 (CD36) secondary to activation of extracellular signal-regulated kinases (ERK)1/2, both of which were abolished by Peptide 19-2.5. Taken together, these results demonstrate that the AMP, Peptide 19-2.5 reduces insulin-resistance, steatohepatitis and proteinuria. These effects are, at least in part, due to prevention of the expression of CD36 and may provide further evidence for a role of metabolic endotoxemia in the pathogenesis of metaflammation and ultimately T2DM. The observed increase in the levels of the endogenous AMP LL-37 in patients with T2DM may serve to limit the severity of the disease.
Collapse
Affiliation(s)
- Shireen Mohammad
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sura Al Zoubi
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Basic Medical Sciences, School of Medicine, Al-Balqa Applied University, As-Salt, Jordan
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Bianka Wissuwa
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | | | - Gareth S. D. Purvis
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Sir William Dunn School Pathology, University of Oxford, Oxford, United Kingdom
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
- Società Italiana per lo Studio della Aterosclerosi (S.I.S.A.) Centre for the Study of Atherosclerosis, Bassini Hospital, Milan, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- IRCCS Multimedica, Sesto San Giovanni, Milan, Italy
- Società Italiana per lo Studio della Aterosclerosi (S.I.S.A.) Centre for the Study of Atherosclerosis, Bassini Hospital, Milan, Italy
| | - Egle Solito
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universitá degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Elisabeth Zechendorf
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Tobias Schürholz
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, c/o Forschungszentrum Borstel, Borstel, Germany
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Muhammad M. Yaqoob
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Lukas Martin
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Thiemermann
- William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
19
|
High-Density Lipoproteins and the Kidney. Cells 2021; 10:cells10040764. [PMID: 33807271 PMCID: PMC8065870 DOI: 10.3390/cells10040764] [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: 03/09/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia is a typical trait of patients with chronic kidney disease (CKD) and it is typically characterized by reduced high-density lipoprotein (HDL)-cholesterol(c) levels. The low HDL-c concentration is the only lipid alteration associated with the progression of renal disease in mild-to-moderate CKD patients. Plasma HDL levels are not only reduced but also characterized by alterations in composition and structure, which are responsible for the loss of atheroprotective functions, like the ability to promote cholesterol efflux from peripheral cells and antioxidant and anti-inflammatory proprieties. The interconnection between HDL and renal function is confirmed by the fact that genetic HDL defects can lead to kidney disease; in fact, mutations in apoA-I, apoE, apoL, and lecithin–cholesterol acyltransferase (LCAT) are associated with the development of renal damage. Genetic LCAT deficiency is the most emblematic case and represents a unique tool to evaluate the impact of alterations in the HDL system on the progression of renal disease. Lipid abnormalities detected in LCAT-deficient carriers mirror the ones observed in CKD patients, which indeed present an acquired LCAT deficiency. In this context, circulating LCAT levels predict CKD progression in individuals at early stages of renal dysfunction and in the general population. This review summarizes the main alterations of HDL in CKD, focusing on the latest update of acquired and genetic LCAT defects associated with the progression of renal disease.
Collapse
|
20
|
A cohort study on risk factors of high-density lipoprotein cholesterol hypolipidemia among urban Chinese adults. Lipids Health Dis 2021; 20:20. [PMID: 33618731 PMCID: PMC7898430 DOI: 10.1186/s12944-021-01449-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/15/2021] [Indexed: 11/17/2022] Open
Abstract
Background High-density lipoprotein cholesterol (HDL-C) hypolipidemia, a major type of dyslipidemia, has been associated with many kinds of diseases, such as stroke, coronary heart disease, obesity and diabetes, and has displayed an increasing prevalence in China. This study explores the risk factors of HDL-C hypolipidemia and makes recommendations for controlling and preventing HDL-C hypolipidemia and the diseases caused by it. Methods Using a retrospective cohort study design, 26,863 urban adults without dyslipidemia, diabetes, cardiovascular and cerebrovascular diseases, hepatosis, renal insufficiency and thyroid diseases were enrolled in the study between 2010 and 2015. Data on each individual were collected at the 2010 baseline year and at a follow-up medical check. A Cox regression model was constructed to evaluate the influence of potential risk factors on the outcome event- HDL-C hypolipidemia. Results The incidence of HDL-C hypolipidemia was 5.7% (1531/26863). Sex, age, body mass index (BMI), HDL-C, triglyceride (TG) and urea nitrogen (UN) were significant risk factors of HDL-C hypolipidemia. Men were more likely to develop HDL-C hypolipidemia than women during follow-up medical checks (HR = 1.258, P = 0.014). The incidence of HDL-C hypolipidemia in the over 65 years old group was higher than that of the ≤65 age group (HR = 1.276, P = 0.009). The incidence of HDL-C hypolipidemia increased with increasing BMI (HR = 1.030, P = 0.002), TG (HR = 1.321, P = 0.001) and UN (HR = 1.054, P = 0.019), while falling with increasing HDL-C in the baseline year (HR = 0.002, P < 0.001). Conclusions Men, aged over 65, with high BMI were at the highest risk of developing HDL-C hypolipidemia. Measures should be taken to prevent HDL-C hypolipidemia even for healthy urban adults whose blood biochemical indicators were in the normal range when their level of TG, UN and HDL-C are closed to the border of the normal value range.
Collapse
|
21
|
Romagnani A, Rottoli E, Mazza EMC, Rezzonico-Jost T, De Ponte Conti B, Proietti M, Perotti M, Civanelli E, Perruzza L, Catapano AL, Baragetti A, Tenedini E, Tagliafico E, Falzoni S, Di Virgilio F, Norata GD, Bicciato S, Grassi F. P2X7 Receptor Activity Limits Accumulation of T Cells within Tumors. Cancer Res 2020; 80:3906-3919. [PMID: 32699136 DOI: 10.1158/0008-5472.can-19-3807] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/01/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
Extracellular ATP (eATP) is a signaling molecule that variably affects all cells of the immune system either directly or after hydrolysis to adenosine. Although eATP is virtually absent in the interstitium of normal tissues, it can be present in the hundreds of micromolar range in tumors, a concentration compatible with activation of the ATP-gated ionotropic P2X7 receptor. Here, we show that P2X7 activity in tumor-infiltrating lymphocytes (TIL) induces cellular senescence and limits tumor suppression. P2X7 stimulation affected cell cycling of effector T cells and resulted in generation of mitochondrial reactive oxygen species and p38 MAPK-dependent upregulation of cyclin-dependent kinase inhibitor 1A (Cdkn1a, encoding for p21Waf1/Cip1). Lack of P2X7 promoted a transcriptional signature that correlated with enhanced cytotoxic T-cell response in human solid tumors. In mice, transfer of tumor-specific T cells with deletion of P2rx7 significantly reduced tumor growth and extended survival. Collectively, these findings uncover a purinergic checkpoint that can be targeted to improve the efficacy of cancer immunotherapy strategies. SIGNIFICANCE: These findings suggest that the purinergic checkpoint P2X7 may be targeted to enhance T-cell-mediated cancer immunotherapy and improve T effector cell accumulation in the tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3906/F1.large.jpg.
Collapse
Affiliation(s)
- Andrea Romagnani
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Elsa Rottoli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | - Tanja Rezzonico-Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Benedetta De Ponte Conti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Michele Proietti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Michela Perotti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland.,Institute for Microbiology, ETH Zürich, Zürich, Switzerland
| | - Elisa Civanelli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alberico L Catapano
- Department of Excellence in Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.,IRCSS Multimedica, Milan, Italy
| | - Andrea Baragetti
- Department of Excellence in Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elena Tenedini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Tagliafico
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Danilo Norata
- Department of Excellence in Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland. .,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi," Milan, Italy
| |
Collapse
|
22
|
Nam KH, Chang TI, Joo YS, Kim J, Lee S, Lee C, Yun HR, Park JT, Yoo TH, Sung SA, Lee KB, Oh KH, Kim SW, Lee J, Kang SW, Choi KH, Ahn C, Han SH. Association Between Serum High-Density Lipoprotein Cholesterol Levels and Progression of Chronic Kidney Disease: Results From the KNOW-CKD. J Am Heart Assoc 2020; 8:e011162. [PMID: 30859896 PMCID: PMC6475054 DOI: 10.1161/jaha.118.011162] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background High-density lipoprotein cholesterol ( HDL -C) levels are generally decreased in patients with chronic kidney disease ( CKD ). However, studies on the relationship between HDL -C and CKD progression are scarce. Methods and Results We studied the association between serum HDL -C levels and the risk of CKD progression in 2168 participants of the KNOW - CKD (Korean Cohort Study for Outcome in Patients With Chronic Kidney Disease). The primary outcome was the composite of a 50% decline in estimated glomerular filtration rate from baseline or end-stage renal disease. The secondary outcome was the onset of end-stage renal disease. During a median follow-up of 3.1 (interquartile range, 1.6-4.5) years, the primary outcome occurred in 335 patients (15.5%). In a fully adjusted Cox model, the lowest category with HDL -C of <30 mg/dL (hazard ratio, 2.21; 95% CI, 1.30-3.77) and the highest category with HDL -C of ≥60 mg/dL (hazard ratio, 2.05; 95% CI , 1.35-3.10) were associated with a significantly higher risk of the composite renal outcome, compared with the reference category with HDL -C of 50 to 59 mg/dL. This association remained unaltered in a time-varying Cox analysis. In addition, a fully adjusted cubic spline model with HDL -C being treated as a continuous variable yielded similar results. Furthermore, consistent findings were obtained in a secondary outcome analysis for the development of end-stage renal disease. Conclusions A U-shaped association was observed between serum HDL -C levels and adverse renal outcomes in this large cohort of patients with CKD . Our findings suggest that both low and high serum HDL -C levels may be detrimental to patients with nondialysis CKD .
Collapse
Affiliation(s)
- Ki Heon Nam
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea.,2 Division of Integrated Medicine Department of Internal Medicine College of Medicine Yonsei University Seoul Korea
| | - Tae Ik Chang
- 4 Department of Internal Medicine National Health Insurance Service Medical Center Ilsan Hospital Goyang-si Gyeonggi-do Korea
| | - Young Su Joo
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Joohwan Kim
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Sangmi Lee
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Changhyun Lee
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Hae-Ryong Yun
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Jung Tak Park
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Tae-Hyun Yoo
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Su Ah Sung
- 5 Department of Internal Medicine Eulji General Hospital Eulji School of Medicine Seoul Korea
| | - Kyu-Beck Lee
- 6 Department of Internal Medicine Sungkyunkwan University School of Medicine Kangbuk Samsung Hospital Seoul Korea
| | - Kook-Hwan Oh
- 7 Department of Internal Medicine Seoul National University Hospital Seoul Korea
| | - Soo Wan Kim
- 8 Department of Internal Medicine Chonnam National University Hospital and Medical School Gwangju Korea
| | - Joongyub Lee
- 9 Department of Prevention and Management Inha University School of Medicine Incheon Korea
| | - Shin-Wook Kang
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea.,3 Department of Internal Medicine College of Medicine Severance Biomedical Science Institute Brain Korea 21 PLUS Yonsei University Seoul Korea
| | - Kyu Hun Choi
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Curie Ahn
- 7 Department of Internal Medicine Seoul National University Hospital Seoul Korea
| | - Seung Hyeok Han
- 1 Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | | |
Collapse
|
23
|
Baragetti A, Ossoli A, Strazzella A, Simonelli S, Baragetti I, Grigore L, Pellegatta F, Catapano AL, Norata GD, Calabresi L. Low Plasma Lecithin: Cholesterol Acyltransferase (LCAT) Concentration Predicts Chronic Kidney Disease. J Clin Med 2020; 9:jcm9072289. [PMID: 32708515 PMCID: PMC7408930 DOI: 10.3390/jcm9072289] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 01/22/2023] Open
Abstract
Low high-density lipoprotein-cholesterol (HDL-c) is the most remarkable lipid trait both in mild-to-moderate chronic kidney disease (CKD) patients as well as in advanced renal disease stages, and we have previously shown that reduced lecithin:cholesterol acyltransferase (LCAT) concentration is a major determinant of the low HDL phenotype. In the present study, we test the hypothesis that reduced LCAT concentration in CKD contributes to the progression of renal damage. The study includes two cohorts of subjects selected from the PLIC study: a cohort of 164 patients with CKD (NefroPLIC cohort) and a cohort of 164 subjects selected from the PLIC participants with a basal estimated glomerular filtration rate (eGFR) > 60 mL/min/1.73 m2 (PLIC cohort). When the NefroPLIC patients were categorized according to the LCAT concentration, patients in the 1st tertile showed the highest event rate at follow-up with an event hazard ratio significantly higher compared to the 3rd LCAT tertile. Moreover, in the PLIC cohort, subjects in the 1st LCAT tertile showed a significantly faster impairment of kidney function compared to subjects in the 3rd LCAT tertile. Serum from subjects in the 1st LCAT tertile promoted a higher reactive oxygen species (ROS) production in renal cells compared to serum from subjects in the third LCAT tertile, and this effect was contrasted by pre-incubation with recombinant human LCAT (rhLCAT). The present study shows that reduced plasma LCAT concentration predicts CKD progression over time in patients with renal dysfunction, and, even more striking, it predicts the impairment of kidney function in the general population.
Collapse
Affiliation(s)
- Andrea Baragetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy;
| | - Alice Ossoli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, 20133 Milano, Italy; (A.O.); (A.S.); (S.S.)
| | - Arianna Strazzella
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, 20133 Milano, Italy; (A.O.); (A.S.); (S.S.)
| | - Sara Simonelli
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, 20133 Milano, Italy; (A.O.); (A.S.); (S.S.)
| | - Ivano Baragetti
- Department of Nephrology and Dialysis, Ospedale Bassini, ASST Nord Milano-Cinisello Balsamo, 20092 Milano, Italy;
| | - Liliana Grigore
- S.I.S.A. Centro per lo Studio della Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, 20092 Milano, Italy; (L.G.); (F.P.)
- IRCCS Ospedale Multimedica, Sesto San Giovanni, 20099 Milano, Italy
| | - Fabio Pellegatta
- S.I.S.A. Centro per lo Studio della Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, 20092 Milano, Italy; (L.G.); (F.P.)
- IRCCS Ospedale Multimedica, Sesto San Giovanni, 20099 Milano, Italy
| | - Alberico L. Catapano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy;
- IRCCS Ospedale Multimedica, Sesto San Giovanni, 20099 Milano, Italy
- Correspondence: (A.L.C.); (G.D.N.); (L.C.); Tel.: +39-0250318302 (A.L.C.); +39-0250318313 (G.D.N.); +39-0250319906 (L.C.); Fax: +39-0250318386 (A.L.C.); +39-0250318386 (G.D.N.); +39-0250319900 (L.C.)
| | - Giuseppe Danilo Norata
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy;
- S.I.S.A. Centro per lo Studio della Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, 20092 Milano, Italy; (L.G.); (F.P.)
- Correspondence: (A.L.C.); (G.D.N.); (L.C.); Tel.: +39-0250318302 (A.L.C.); +39-0250318313 (G.D.N.); +39-0250319906 (L.C.); Fax: +39-0250318386 (A.L.C.); +39-0250318386 (G.D.N.); +39-0250319900 (L.C.)
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, 20133 Milano, Italy; (A.O.); (A.S.); (S.S.)
- Correspondence: (A.L.C.); (G.D.N.); (L.C.); Tel.: +39-0250318302 (A.L.C.); +39-0250318313 (G.D.N.); +39-0250319906 (L.C.); Fax: +39-0250318386 (A.L.C.); +39-0250318386 (G.D.N.); +39-0250319900 (L.C.)
| |
Collapse
|
24
|
Tan KCB, Cheung CL, Lee ACH, Lam JKY, Wong Y, Shiu SWM. Carbamylated Lipoproteins and Progression of Diabetic Kidney Disease. Clin J Am Soc Nephrol 2020; 15:359-366. [PMID: 32075807 PMCID: PMC7057307 DOI: 10.2215/cjn.11710919] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/07/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND OBJECTIVES Protein carbamylation is a consequence of uremia and carbamylated lipoproteins contribute to atherogenesis in CKD. Proteins can also be carbamylated by a urea-independent mechanism, and whether carbamylated lipoproteins contribute to the progression of CKD has not been investigated. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A case-control study was performed to determine whether there were changes in plasma levels of carbamylated lipoproteins in individuals with type 2 diabetes with eGFR >60 ml/min per 1.73 m2 compared with a group of age- and sex-matched healthy controls. A cohort of 1320 patients with type 2 diabetes with baseline eGFR ≥30 ml/min per 1.73 m2 was longitudinally followed up to evaluate the association between carbamylated lipoproteins and progression of CKD. The primary kidney outcome was defined as doubling of serum creatinine and/or initiation of KRT during follow-up. Plasma carbamylated LDLs and HDLs was measured by ELISA. RESULTS In individuals with diabetes with eGFR >60 ml/min per 1.73 m2, both plasma carbamylated LDL and HDL levels were higher compared with healthy controls (P<0.001). After a mean follow-up of 9 years of the diabetic cohort, individuals in the top quartile of carbamylated LDL (hazard ratio, 2.21; 95% confidence interval, 1.42 to 3.46; P<0.001) and carbamylated HDL (hazard ratio, 4.53; 95% confidence interval, 2.87 to 7.13; P<0.001) had higher risk of deterioration of kidney function compared with those in the lowest quartile. On multivariable Cox regression analysis, plasma carbamylated LDL was no longer associated with kidney outcome after adjusting for baseline eGFR and potential confounding factors. However, the association between plasma carbamylated HDL and kidney outcome remained significant and was independent of HDL cholesterol. CONCLUSIONS Plasma carbamylated HDL but not carbamylated LDL was independently associated with progression of CKD in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Kathryn C B Tan
- Department of Medicine, University of Hong Kong, Hong Kong, China and
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Alan C H Lee
- Department of Medicine, University of Hong Kong, Hong Kong, China and
| | - Joanne K Y Lam
- Department of Medicine, University of Hong Kong, Hong Kong, China and
| | - Ying Wong
- Department of Medicine, University of Hong Kong, Hong Kong, China and
| | - Sammy W M Shiu
- Department of Medicine, University of Hong Kong, Hong Kong, China and
| |
Collapse
|
25
|
Bonacina F, Pirillo A, Catapano AL, Norata GD. Cholesterol membrane content has a ubiquitous evolutionary function in immune cell activation: the role of HDL. Curr Opin Lipidol 2019; 30:462-469. [PMID: 31577612 DOI: 10.1097/mol.0000000000000642] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Cellular cholesterol content influences the structure and function of lipid rafts, plasma membrane microdomains essential for cell signaling and activation. HDL modulate cellular cholesterol efflux, thus limiting cholesterol accumulation and controlling immune cell activation. Aim of this review is to discuss the link between HDL and cellular cholesterol metabolism in immune cells and the therapeutic potential of targeting cholesterol removal from cell membranes. RECENT FINDINGS The inverse relationship between HDL-cholesterol (HDL-C) levels and the risk of cardiovascular disease has been recently challenged by observations linking elevated levels of HDL-C with increased risk of all-cause mortality, infections and autoimmune diseases, paralleled by the failure of clinical trials with HDL-C-raising therapies. These findings suggest that improving HDL function might be more important than merely raising HDL-C levels. New approaches aimed at increasing the ability of HDL to remove cellular cholesterol have been assessed for their effect on immune cells, and the results have suggested that this could be a new effective approach. SUMMARY Cholesterol removal from plasma membrane by different means affects the activity of immune cells, suggesting that approaches aimed at increasing the ability of HDL to mobilize cholesterol from cells would represent the next step in HDL biology.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital
- IRCCS MultiMedica, Milan, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan
- IRCCS MultiMedica, Milan, Italy
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan
- Center for the Study of Atherosclerosis, E. Bassini Hospital
| |
Collapse
|
26
|
Lunasin Improves the LDL-C Lowering Efficacy of Simvastatin via Inhibiting PCSK9 Expression in Hepatocytes and ApoE -/- Mice. Molecules 2019; 24:molecules24224140. [PMID: 31731717 PMCID: PMC6891362 DOI: 10.3390/molecules24224140] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Statins are the most popular therapeutic drugs to lower plasma low density lipoprotein cholesterol (LDL-C) synthesis by competitively inhibiting hydroxyl-3-methyl-glutaryl-CoA (HMG-CoA) reductase and up-regulating the hepatic low density lipoprotein receptor (LDLR). However, the concomitant up-regulation of proprotein convertase subtilisin/kexin type 9 (PCSK9) by statin attenuates its cholesterol lowering efficacy. Lunasin, a soybean derived 43-amino acid polypeptide, has been previously shown to functionally enhance LDL uptake via down-regulating PCSK9 and up-regulating LDLR in hepatocytes and mice. Herein, we investigated the LDL-C lowering efficacy of simvastatin combined with lunasin. In HepG2 cells, after co-treatment with 1 μM simvastatin and 5 μM lunasin for 24 h, the up-regulation of PCSK9 by simvastatin was effectively counteracted by lunasin via down-regulating hepatocyte nuclear factor 1α (HNF-1α), and the functional LDL uptake was additively enhanced. Additionally, after combined therapy with simvastatin and lunasin for four weeks, ApoE−/− mice had significantly lower PCSK9 and higher LDLR levels in hepatic tissues and remarkably reduced plasma concentrations of total cholesterol (TC) and LDL-C, as compared to each monotherapy. Conclusively, lunasin significantly improved the LDL-C lowering efficacy of simvastatin by counteracting simvastatin induced elevation of PCSK9 in hepatocytes and ApoE−/− mice. Simvastatin combined with lunasin could be a novel regimen for hypercholesterolemia treatment.
Collapse
|
27
|
Kidney as modulator and target of "good/bad" HDL. Pediatr Nephrol 2019; 34:1683-1695. [PMID: 30291429 PMCID: PMC6450786 DOI: 10.1007/s00467-018-4104-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 10/28/2022]
Abstract
The strong inverse relationship between low levels of high-density lipoproteins (HDLs) and atherosclerotic cardiovascular disease (CVD) led to the designation of HDL as the "good" cholesterol. The atheroprotection is thought to reflect HDL's capacity to efflux cholesterol from macrophages, followed by interaction with other lipoproteins in the plasma, processing by the liver and excretion into bile. However, pharmacologic increases in HDL-C levels have not led to expected clinical benefits, giving rise to the concept of dysfunctional HDL, in which increases in serum HDL-C are not beneficial due to lost or altered HDL functions and transition to "bad" HDL. It is now understood that the cholesterol in HDL, measured by HDL-C, is neither a marker nor the mediator of HDL function, including cholesterol efflux capacity. It is also understood that besides cholesterol efflux, HDL functionality encompasses many other potentially beneficial functions, including antioxidant, anti-inflammatory, antithrombotic, anti-apoptotic, and vascular protective effects that may be critical protective pathways for various cells, including those in the kidney parenchyma. This review highlights advances in our understanding of the role kidneys play in HDL metabolism, including the effects on levels, composition, and functionality of HDL particles, particularly the main HDL protein, apolipoprotein AI (apoAI). We suggest that normal apoAI/HDL in the glomerular filtrate provides beneficial effects, including lymphangiogenesis, that promote resorption of renal interstitial fluid and biological particles. In contrast, dysfunctional apoAI/HDL activates detrimental pathways in tubular epithelial cells and lymphatics that lead to interstitial accumulation of fluid and harmful particles that promote progressive kidney damage.
Collapse
|
28
|
Inflammatory Dietary Pattern Predicts Dyslipidemia and Anemia in Middle-Aged and Older Taiwanese Adults with Declined Kidney Function: A Cross-Sectional Population Study from 2008 to 2010. Nutrients 2019; 11:nu11092052. [PMID: 31480674 PMCID: PMC6770658 DOI: 10.3390/nu11092052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/17/2019] [Accepted: 08/25/2019] [Indexed: 02/06/2023] Open
Abstract
Dyslipidemia, anemia, and inflammation are associated with declined kidney function. This study investigated the association of inflammatory dietary pattern with dyslipidemia, anemia, and kidney function biomarkers among middle-aged and older Taiwanese adults with declined kidney function. Biochemical data and food frequency questionnaire were obtained from 41,128 participants with estimated glomerular filtration rate (eGFR) <90 mL/min/1.73 m2 and positive urinary protein. Inflammatory dietary pattern was identified by reduced rank regression with C-reactive protein (CRP) and neutrophil-to-lymphocyte ratio (N/L) as response variables. Males had higher prevalence of dyslipidemia and higher inflammatory markers, but lower prevalence of anemia and lower eGFR levels compared to females. Inflammatory dietary pattern characterized with low intakes of seafood, grains, vegetables, and fruits but high intakes of meat, eggs, preserved/processed foods, and sugary drinks was associated with an increased risk of dyslipidemia by 21% in males and an increased risk of anemia by 28–47% in both genders. Furthermore, high consumption of inflammatory dietary pattern was associated with reduced eGFR (males β = −0.85, 95% CI −1.26 to −0.43, females β = −0.53, 95% CI −0.98 to −0.08) and increased N/L and/or CRP in both genders. In conclusion, inflammatory dietary pattern is positively associated with dyslipidemia, anemia, and decreased kidney function in middle-aged and older adults with declined kidney function.
Collapse
|
29
|
Holmes MV, Davey Smith G. Challenges in Interpreting Multivariable Mendelian Randomization: Might "Good Cholesterol" Be Good After All? Am J Kidney Dis 2019; 71:149-153. [PMID: 29389382 DOI: 10.1053/j.ajkd.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/11/2017] [Indexed: 01/29/2023]
Affiliation(s)
- Michael V Holmes
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom; Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom.
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
30
|
Pirillo A, Catapano AL, Norata GD. Biological Consequences of Dysfunctional HDL. Curr Med Chem 2019; 26:1644-1664. [PMID: 29848265 DOI: 10.2174/0929867325666180530110543] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/25/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
Epidemiological studies have suggested an inverse correlation between high-density lipoprotein (HDL) cholesterol levels and the risk of cardiovascular disease. HDLs promote reverse cholesterol transport (RCT) and possess several putative atheroprotective functions, associated to the anti-inflammatory, anti-thrombotic and anti-oxidant properties as well as to the ability to support endothelial physiology. The assumption that increasing HDL-C levels would be beneficial on cardiovascular disease (CVD), however, has been questioned as, in most clinical trials, HDL-C-raising therapies did not result in improved cardiovascular outcomes. These findings, together with the observations from Mendelian randomization studies showing that polymorphisms mainly or solely associated with increased HDL-C levels did not decrease the risk of myocardial infarction, shift the focus from HDL-C levels toward HDL functional properties. Indeed, HDL from atherosclerotic patients not only exhibit impaired atheroprotective functions but also acquire pro-atherogenic properties and are referred to as "dysfunctional" HDL; this occurs even in the presence of normal or elevated HDL-C levels. Pharmacological approaches aimed at restoring HDL functions may therefore impact more significantly on CVD outcome than drugs used so far to increase HDL-C levels. The aim of this review is to discuss the pathological conditions leading to the formation of dysfunctional HDL and their role in atherosclerosis and beyond.
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,IRCCS Multimedica, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia
| |
Collapse
|
31
|
Purvis GSD, Collino M, Loiola RA, Baragetti A, Chiazza F, Brovelli M, Sheikh MH, Collotta D, Cento A, Mastrocola R, Aragno M, Cutrin JC, Reutelingsperger C, Grigore L, Catapano AL, Yaqoob MM, Norata GD, Solito E, Thiemermann C. Identification of AnnexinA1 as an Endogenous Regulator of RhoA, and Its Role in the Pathophysiology and Experimental Therapy of Type-2 Diabetes. Front Immunol 2019; 10:571. [PMID: 30972066 PMCID: PMC6446914 DOI: 10.3389/fimmu.2019.00571] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Annexin A1 (ANXA1) is an endogenously produced anti-inflammatory protein, which plays an important role in the pathophysiology of diseases associated with chronic inflammation. We demonstrate that patients with type-2 diabetes have increased plasma levels of ANXA1 when compared to normoglycemic subjects. Plasma ANXA1 positively correlated with fatty liver index and elevated plasma cholesterol in patients with type-2 diabetes, suggesting a link between aberrant lipid handling, and ANXA1. Using a murine model of high fat diet (HFD)-induced insulin resistance, we then investigated (a) the role of endogenous ANXA1 in the pathophysiology of HFD-induced insulin resistance using ANXA1−/− mice, and (b) the potential use of hrANXA1 as a new therapeutic approach for experimental diabetes and its microvascular complications. We demonstrate that: (1) ANXA1−/− mice fed a HFD have a more severe diabetic phenotype (e.g., more severe dyslipidemia, insulin resistance, hepatosteatosis, and proteinuria) compared to WT mice fed a HFD; (2) treatment of WT-mice fed a HFD with hrANXA1 attenuated the development of insulin resistance, hepatosteatosis and proteinuria. We demonstrate here for the first time that ANXA1−/− mice have constitutively activated RhoA. Interestingly, diabetic mice, which have reduced tissue expression of ANXA1, also have activated RhoA. Treatment of HFD-mice with hrANXA1 restored tissue levels of ANXA1 and inhibited RhoA activity, which, in turn, resulted in restoration of the activities of Akt, GSK-3β and endothelial nitric oxide synthase (eNOS) secondary to re-sensitization of IRS-1 signaling. We further demonstrate in human hepatocytes that ANXA1 protects against excessive mitochondrial proton leak by activating FPR2 under hyperglycaemic conditions. In summary, our data suggest that (a) ANXA1 is a key regulator of RhoA activity, which restores IRS-1 signal transduction and (b) recombinant human ANXA1 may represent a novel candidate for the treatment of T2D and/or its complications.
Collapse
Affiliation(s)
- Gareth S D Purvis
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Rodrigo A Loiola
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Martina Brovelli
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy.,Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy
| | - Madeeha H Sheikh
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Alessia Cento
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Raffaella Mastrocola
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Manuela Aragno
- Department of Molecular Biotechnology and Sciences for the Health, University of Turin, Turin, Italy
| | - Juan C Cutrin
- Department of Molecular Biotechnology and Sciences for the Health, University of Turin, Turin, Italy
| | - Chris Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute, Maastricht University, Maastricht, Netherlands
| | - Liliana Grigore
- Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy.,IRCCS Multimedica, Lombardy, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Magdi M Yaqoob
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy.,Centro SISA per lo studio del'Aterosclerosi, Bassini Hospital, Lombardy, Italy
| | - Egle Solito
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - Christoph Thiemermann
- Department of Translational Medicine and Therapeutics, Bart's and The London School of Medicine and Dentistry, The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
32
|
Gille A, Duffy D, Tortorici MA, Wright SD, Deckelbaum LI, D'Andrea DM. Moderate Renal Impairment Does Not Impact the Ability of CSL112 (Apolipoprotein A-I [Human]) to Enhance Cholesterol Efflux Capacity. J Clin Pharmacol 2018; 59:427-436. [PMID: 30452776 PMCID: PMC6587782 DOI: 10.1002/jcph.1337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/17/2018] [Indexed: 12/31/2022]
Abstract
CSL112 (apolipoprotein A‐I [human]) is a novel intravenous formulation of plasma‐derived apolipoprotein A‐I (apoA‐I) that enhances cholesterol efflux capacity. Renal impairment is a common comorbidity in acute myocardial infarction patients and is associated with impaired lipid metabolism. The aim of this phase 1 study was to assess the impact of moderate renal impairment on the pharmacokinetic and pharmacodynamic profile of CSL112. Sixteen subjects with moderate renal impairment and 16 age‐, sex‐, and weight‐matched subjects with normal renal function participated in the study. Within each renal function cohort, subjects were randomized 3:1 to receive a single intravenous infusion of CSL112 2 g (n = 6) or placebo (n = 2) or CSL112 6 g (n = 6) or placebo (n = 2). At baseline, subjects with moderate renal impairment versus normal renal function had higher total cholesterol efflux, ABCA1‐dependent cholesterol efflux capacity, and pre‐β1‐high‐density lipoprotein (HDL) levels. Infusing CSL112 resulted in similar, immediate, robust, dose‐dependent elevations in apoA‐I and cholesterol efflux capacity in both renal function cohorts and significantly greater elevations in pre‐β1‐HDL (P < .05) in moderate renal impairment. Lecithin‐cholesterol acyltransferase activity, demonstrated by a time‐dependent change in the ratio of unesterified to esterified cholesterol, did not differ by renal function. No meaningful changes in proatherogenic lipid levels were observed. Moderate renal impairment did not impact the ability of CSL112 to enhance cholesterol efflux capacity. CSL112 may represent a novel therapy to reduce the risk of early recurrent cardiovascular events following acute myocardial infarction in patients with or without moderate renal impairment.
Collapse
|
33
|
Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat Commun 2018; 9:3083. [PMID: 30082772 PMCID: PMC6079066 DOI: 10.1038/s41467-018-05322-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 06/24/2018] [Indexed: 01/06/2023] Open
Abstract
Cholesterol homeostasis has a pivotal function in regulating immune cells. Here we show that apolipoprotein E (apoE) deficiency leads to the accumulation of cholesterol in the cell membrane of dendritic cells (DC), resulting in enhanced MHC-II-dependent antigen presentation and CD4+ T-cell activation. Results from WT and apoE KO bone marrow chimera suggest that apoE from cells of hematopoietic origin has immunomodulatory functions, regardless of the onset of hypercholesterolemia. Humans expressing apoE4 isoform (ε4/3–ε4/4) have increased circulating levels of activated T cells compared to those expressing WT apoE3 (ε3/3) or apoE2 isoform (ε2/3–ε2/2). This increase is caused by enhanced antigen-presentation by apoE4-expressing DCs, and is reversed when these DCs are incubated with serum containing WT apoE3. In summary, our study identifies myeloid-produced apoE as a key physiological modulator of DC antigen presentation function, paving the way for further explorations of apoE as a tool to improve the management of immune diseases. Cholesterol homeostasis can modulate immunity via multiple pathways. Here the authors show that apolipoprotein E, an important regulator of cholesterol, produced by myeloid cells can regulate T cell activation by controlling the antigen presentation activity of dendritic cells in both humans and mice.
Collapse
|
34
|
Estrella MM, Parekh RS. The Expanding Role of APOL1 Risk in Chronic Kidney Disease and Cardiovascular Disease. Semin Nephrol 2018; 37:520-529. [PMID: 29110759 DOI: 10.1016/j.semnephrol.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Variants of the APOL1 gene, found primarily in individuals of African descent, are associated with various forms of kidney disease and kidney disease progression. Recent studies evaluating the association of APOL1 with cardiovascular disease have yielded conflicting results, and the potential role in cardiovascular disease remains unclear. In this review, we summarize the observational studies linking the APOL1 risk variants with chronic kidney and cardiovascular disease among persons of African descent.
Collapse
Affiliation(s)
- Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California San Francisco, San Francisco, CA; Department of Medicine, San Francisco VA Medical Center, San Francisco, CA
| | - Rulan S Parekh
- Division of Nephrology, Departments of Pediatrics and Medicine, The Hospital for Sick Children, SickKids Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Kaseda R, Tsuchida Y, Yang HC, Yancey PG, Zhong J, Tao H, Bian A, Fogo AB, Linton MRF, Fazio S, Ikizler TA, Kon V. Chronic kidney disease alters lipid trafficking and inflammatory responses in macrophages: effects of liver X receptor agonism. BMC Nephrol 2018; 19:17. [PMID: 29374468 PMCID: PMC5787279 DOI: 10.1186/s12882-018-0814-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/15/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Our aim was to evaluate lipid trafficking and inflammatory response of macrophages exposed to lipoproteins from subjects with moderate to severe chronic kidney disease (CKD), and to investigate the potential benefits of activating cellular cholesterol transporters via liver X receptor (LXR) agonism. METHODS LDL and HDL were isolated by sequential density gradient ultracentrifugation of plasma from patients with stage 3-4 CKD and individuals without kidney disease (HDLCKD and HDLCont, respectively). Uptake of LDL, cholesterol efflux to HDL, and cellular inflammatory responses were assessed in human THP-1 cells. HDL effects on inflammatory markers (MCP-1, TNF-α, IL-1β), Toll-like receptors-2 (TLR-2) and - 4 (TLR-4), ATP-binding cassette class A transporter (ABCA1), NF-κB, extracellular signal regulated protein kinases 1/2 (ERK1/2) were assessed by RT-PCR and western blot before and after in vitro treatment with an LXR agonist. RESULTS There was no difference in macrophage uptake of LDL isolated from CKD versus controls. By contrast, HDCKD was significantly less effective than HDLCont in accepting cholesterol from cholesterol-enriched macrophages (median 20.8% [IQR 16.1-23.7] vs control (26.5% [IQR 19.6-28.5]; p = 0.008). LXR agonist upregulated ABCA1 expression and increased cholesterol efflux to HDL of both normal and CKD subjects, although the latter continued to show lower efflux capacity. HDLCKD increased macrophage cytokine response (TNF-α, MCP-1, IL-1β, and NF-κB) versus HDLCont. The heightened cytokine response to HDLCKD was further amplified in cells treated with LXR agonist. The LXR-augmentation of inflammation was associated with increased TLR-2 and TLR-4 and ERK1/2. CONCLUSIONS Moderate to severe impairment in kidney function promotes foam cell formation that reflects impairment in cholesterol acceptor function of HDLCKD. Activation of cellular cholesterol transporters by LXR agonism improves but does not normalize efflux to HDLCKD. However, LXR agonism actually increases the pro-inflammatory effects of HDLCKD through activation of TLRs and ERK1/2 pathways.
Collapse
Affiliation(s)
- Ryohei Kaseda
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Yohei Tsuchida
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Hai-Chun Yang
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
| | | | - Jianyong Zhong
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| | - Huan Tao
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Aihua Bian
- Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Agnes B. Fogo
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Mac Rae F. Linton
- Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Pharmacology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Oregon, Portland USA
| | | | - Valentina Kon
- Departments of Pediatrics, Vanderbilt University Medical Center, 1161 21st Avenue South, C-4204 Medical Center North, Nashville, TN 37232-2584 USA
| |
Collapse
|
36
|
Baragetti A, Ramirez GA, Magnoni M, Garlaschelli K, Grigore L, Berteotti M, Scotti I, Bozzolo E, Berti A, Camici PG, Catapano AL, Manfredi AA, Ammirati E, Norata GD. Disease trends over time and CD4 +CCR5 + T-cells expansion predict carotid atherosclerosis development in patients with systemic lupus erythematosus. Nutr Metab Cardiovasc Dis 2018; 28:53-63. [PMID: 29150407 DOI: 10.1016/j.numecd.2017.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 09/07/2017] [Accepted: 09/09/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Patients with Systemic Lupus Erythematosus (SLE) present increased cardiovascular mortality compared to the general population. Few studies have assessed the long-term development and progression of carotid atherosclerotic plaque in SLE patients. Our aim was to investigate the association of clinical and laboratory markers of disease activity and classical cardiovascular risk factors (CVRF) with carotid atherosclerosis development in SLE patients in a prospective 5-year study. METHODS AND RESULTS Clinical history and information on principal CVRFs were collected at baseline and after 5 years in 40 SLE patients (36 women, mean age 42 ± 9 years; 14.4 ± 7 years of mean disease duration) and 50 age-matched controls. Carotid Doppler ultrasonography was employed to quantify the atherosclerotic burden at baseline and at follow up. Clinimetrics were applied to assess SLE activity over time (SLEDAI). The association between basal circulating T cell subsets (including CD4+CCR5+; CD4+CXCR3+; CD4+HLADR+; CD4+CD45RA+RO-, CD4+CD45RO+RA- and their subsets) and atherosclerosis development was evaluated. During the 5-year follow up, 32% of SLE patients, developed carotid atherosclerosis compared to 4% of controls. Furthermore, considering SLEDAI changes over time, patients within the highest tertile were those with increased incidence of carotid atherosclerosis independently of CVRF. In addition, increased levels of CD4+CCR5+ T cells were independently associated with the development of carotid atherosclerosis in SLE patients. CONCLUSION Serial clinical evaluations over time, rather than a single point estimation of disease activity or CVRF burden, are required to define the risk of carotid atherosclerosis development in SLE patients. Specific T cell subsets are associated with long-term atherosclerotic progression and may further be of help in predicting vascular disease progression.
Collapse
Affiliation(s)
- A Baragetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy
| | - G A Ramirez
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Magnoni
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - K Garlaschelli
- Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy
| | - L Grigore
- Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy; IRCCS - Multimedica Hospital, Sesto San Giovanni, Italy
| | - M Berteotti
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - I Scotti
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - E Bozzolo
- Università Vita-Salute San Raffaele, Milan, Italy
| | - A Berti
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - P G Camici
- Department of Thoracic and Cardiovascular Surgery, Università Vita-Salute San Raffaele Scientific Institute Milan, Italy
| | - A L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; IRCCS - Multimedica Hospital, Sesto San Giovanni, Italy
| | - A A Manfredi
- Università Vita-Salute San Raffaele, Milan, Italy; Unit of Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - E Ammirati
- Niguarda Ca' Granda Hospital, Milan, Italy
| | - G D Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy; Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Perth, Western Australia, Australia.
| |
Collapse
|
37
|
Wen J, Chen Y, Huang Y, Lu Y, Liu X, Zhou H, Yuan H. Association of the TG/HDL-C and Non-HDL-C/HDL-C Ratios with Chronic Kidney Disease in an Adult Chinese Population. Kidney Blood Press Res 2017; 42:1141-1154. [PMID: 29224024 DOI: 10.1159/000485861] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/23/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Evidence indicates a role for dyslipidemia in the development of chronic kidney disease (CKD). However, the association of lipid abnormalities and their ratios with kidney disease using the new CKD Epidemiology Collaboration (CKD-EPI) equation is not well understood. METHODS This cross-sectional study included 48,054 adult subjects. CKD was defined as an estimated glomerular filtration rate <60 ml/min/1.73 m2 or dipstick-positive proteinuria. Logistic regression models were used to examine the relationship between lipid variables and CKD. RESULTS The prevalence of CKD in this study was 3.7%. When the participants exhibited higher serum triglyceride (TG), a higher TG/high-density lipoprotein cholesterol (TG/HDL-c) ratio or a higher non-HDL-c/HDL-c ratio or HDL-c in a lower quartile, the prevalence of CKD tended to be higher. The multivariate adjusted odds ratios for CKD per 1 standard deviation increase in lipid level were 1.17 (1.10-1.23) for TG, 0.86 (0.79-0.93) for HDL-c, 1.21 (1.13-1.31) for the TG/HDL-c ratio, and 1.14 (1.06-1.22) for the non-HDL-c/HDL-c ratio. No significant association was detected between CKD and total cholesterol (TC), non-HDL-c or the low-density lipoprotein cholesterol/HDL-c (LDL-c/HDL-c) ratio. CONCLUSION In this relatively healthy adult Chinese population, the CKD-EPI equation determined that the TG/HDL-c and non-HDL-c/HDL-c ratios as well as TG and HDL-c correlate with the prevalence of CKD.
Collapse
Affiliation(s)
- Jia Wen
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Center of Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yiyin Chen
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yun Huang
- Pharmacy Department of Ningbo City Medical Treatment Center, Lihuili Hospital, Ningbo, China
| | - Yao Lu
- Center of Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xing Liu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Center of Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Honghao Zhou
- Institute of Clinical Pharmacology of Central South University, Changsha, China
| | - Hong Yuan
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Center of Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
38
|
Yin P, Zhou Y, Li B, Hong L, Chen W, Yu X. Effect of low and high HDL-C levels on the prognosis of lupus nephritis patients: a prospective cohort study. Lipids Health Dis 2017; 16:232. [PMID: 29212518 PMCID: PMC5719733 DOI: 10.1186/s12944-017-0622-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/22/2017] [Indexed: 11/16/2022] Open
Abstract
Background Few data has been available on the effect of serum HDL-C levels on the prognosis of lupus nephritis (LN) patients. The present study therefore aimed to explore the effect of serum HDL-C levels on LN patients. Methods We included 775 patients with follow-up information registered in an LN database between 1 January 2006 and 31 December 2011. The patients were divided into groups with low, intermediate and high HDL-C, according to NCEP ATPIII criteria. Cox regression analyses were used to explore the effects of HDL-C levels on end-stage renal disease (ESRD), all-cause mortality and cardiovascular disease (CVD) mortality. Results During a median follow-up of 56 months (3–206 months), 71 (9.2%) had ESRD. 84 (10.8%) deaths occurred, 17 (20.2%) of which were due to CVD. There was no statistically significant association of HDL-C category or continuous HDL-C levels with ESRD in the total cohort, but in subgroup analyses by eGFR, with each 0.1 mmol/L increase in HDL-C level, adjusted HRs for ESRD were 0.92 (95% CI: 0.83–1.04, P = 0.173) for eGFR ≥60 ml/min/1.73m2 and 1.11 (95% CI: 1.01–1.23, P = 0.036) for eGFR <60 ml/min/1.73m2. The effect of the interaction between eGFR category and serum HDL-C level on ESRD was statistically significant (β = −1.738, P = 0.005). Low HDL-C was associated with all-cause mortality (HR = 2.16, 95% CI: 1.06–4.40, P = 0.033) with intermediate HDL-C as reference category after adjusting for several variables. Conclusions Our results demonstrate that high HDL-C levels were associated with increased risk of ESRD in LN patients with advanced renal dysfunction. While low HDL-C levels were associated with increased risk of all-cause mortality in LN patients. Trial registration ClinicalTrials.gov Identifier: NCT03001973, 22 December 2016 retrospectively registered. Electronic supplementary material The online version of this article (10.1186/s12944-017-0622-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peiran Yin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Ying Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Bin Li
- Clinical Research Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lingyao Hong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China. .,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China.
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China.,Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
39
|
Zinellu A, Sotgia S, Sotgiu E, Assaretti S, Baralla A, Mangoni AA, Satta AE, Carru C. Cholesterol lowering treatment restores blood global DNA methylation in chronic kidney disease (CKD) patients. Nutr Metab Cardiovasc Dis 2017; 27:822-829. [PMID: 28755807 DOI: 10.1016/j.numecd.2017.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/19/2017] [Accepted: 06/19/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Chronic kidney disease (CKD) is characterized by increased oxidative stress (OS). In consideration of the well-known link between OS and DNA methylation we assessed DNA methylcytosine (mCyt) concentrations in CKD patients at baseline and during cholesterol lowering treatment. METHODS AND RESULTS DNA methylation and OS indices (malonyldialdehyde, MDA; allantoin/uric acid ratio, All/UA) were measured in 30 CKD patients randomized to three cholesterol lowering regimens for 12 months (simvastatin 40 mg/day, ezetimibe/simvastatin 10/20 mg/day, or ezetimibe/simvastatin 10/40 mg/day) and 30 age- and sex-matched healthy controls. DNA methylation was significantly lower in CKD patients vs. controls (4.06 ± 0.20% vs. 4.27 ± 0.17% mCyt, p = 0.0001). Treatment significantly increased mCyt DNA concentrations in all patients (4.06 ± 0.04% at baseline; 4.12 ± 0.03% at 4 months; 4.17 ± 0.03% at 8 months; and 4.20 ± 0.02% at 12 months, p = 0.0001 for trend). A trend for a greater effect on DNA methylation was observed with combined treatment ezetimibe/simvastatin 10/40 mg/day (+5.2% after one year treatment). The treatment-associated mCyt increase was significantly correlated with the concomitant reduction in MDA concentrations and All/AU ratios. CONCLUSION Our results demonstrate that CKD patients have a lower degree of DNA methylation and that cholesterol lowering treatment restores mCyt DNA concentrations to levels similar to healthy controls. The treatment-associated increase in DNA methylation is correlated with a concomitant reduction in OS markers. The study was registered at clinicaltrials.gov (NCT00861731).
Collapse
Affiliation(s)
- A Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| | - S Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - E Sotgiu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - S Assaretti
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - A Baralla
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - A A Mangoni
- Department of Clinical Pharmacology, School of Medicine, Flinders University, Adelaide, Australia
| | - A E Satta
- Department of Surgical, Microsurgical and Medical Sciences, University of Sassari, Sassari, Italy
| | - C Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital Sassari (AOU), Sassari, Italy
| |
Collapse
|
40
|
Baragetti A, Balzarotti G, Grigore L, Pellegatta F, Guerrini U, Pisano G, Fracanzani AL, Fargion S, Norata GD, Catapano AL. PCSK9 deficiency results in increased ectopic fat accumulation in experimental models and in humans. Eur J Prev Cardiol 2017; 24:1870-1877. [DOI: 10.1177/2047487317724342] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Italy
| | - Gloria Balzarotti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Liliana Grigore
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Italy
- IRCCS Multimedica Hospital, Italy
| | - Fabio Pellegatta
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Italy
- IRCCS Multimedica Hospital, Italy
| | - Uliano Guerrini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Giuseppina Pisano
- Department of Pathophysiology and Transplantation, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital, Università degli Studi di Milano, Italy
| | - Anna L Fracanzani
- Department of Pathophysiology and Transplantation, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital, Università degli Studi di Milano, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital, Università degli Studi di Milano, Italy
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Western Australia
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
- IRCCS Multimedica Hospital, Italy
| |
Collapse
|
41
|
de Gonzalo-Calvo D, Cenarro A, Garlaschelli K, Pellegatta F, Vilades D, Nasarre L, Camino-Lopez S, Crespo J, Carreras F, Leta R, Catapano AL, Norata GD, Civeira F, Llorente-Cortes V. Translating the microRNA signature of microvesicles derived from human coronary artery smooth muscle cells in patients with familial hypercholesterolemia and coronary artery disease. J Mol Cell Cardiol 2017; 106:55-67. [PMID: 28342976 DOI: 10.1016/j.yjmcc.2017.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/17/2022]
Abstract
AIMS To analyze the impact of atherogenic lipoproteins on the miRNA signature of microvesicles derived from human coronary artery smooth muscle cells (CASMC) and to translate these results to familial hypercholesterolemia (FH) and coronary artery disease (CAD) patients. METHODS Conditioned media was collected after exposure of CASMC to atherogenic lipoproteins. Plasma samples were collected from two independent populations of diagnosed FH patients and matched normocholesterolemic controls (Study population 1, N=50; Study population 2, N=24) and a population of patients with suspected CAD (Study population 3, N=50). Extracellular vesicles were isolated and characterized using standard techniques. A panel of 30 miRNAs related to vascular smooth muscle cell (VSMC) (patho-)physiology was analyzed using RT-qPCR. RESULTS Atherogenic lipoproteins significantly reduced levels of miR-15b-5p, -24-3p, -29b-3p, -130a-3p, -143-3p, -146a-3p, -222-3p, -663a levels (P<0.050) in microvesicles (0.1μm-1μm in diameter) released by CASMC. Two of these miRNAs, miR-24-3p and miR-130a-3p, were reduced in circulating microvesicles from FH patients compared with normocholesterolemic controls in a pilot study (Study population 1) and in different validation studies (Study populations 1 and 2) (P<0.050). Supporting these results, plasma levels of miR-24-3p and miR-130a-3p were also downregulated in FH patients compared to controls (P<0.050). In addition, plasma levels of miR-130a-3p were inversely associated with coronary atherosclerosis in a cohort of suspected CAD patients (Study population 3) (P<0.050). CONCLUSIONS Exposure to atherogenic lipoproteins modifies the miRNA profile of CASMC-derived microvesicles and these alterations are reflected in patients with FH. Circulating miR-130a-3p emerges as a potential biomarker for coronary atherosclerosis.
Collapse
Affiliation(s)
- David de Gonzalo-Calvo
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.
| | - Ana Cenarro
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Lipid Unit and Molecular Research Laboratory, IIS Aragón, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, Spain
| | - Katia Garlaschelli
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy
| | - Fabio Pellegatta
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy; IRCCS Multimedica, Milan, Italy
| | - David Vilades
- Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Laura Nasarre
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sandra Camino-Lopez
- Catalan Institute of Cardiovascular Sciences, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Javier Crespo
- Catalan Institute of Cardiovascular Sciences, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Francesc Carreras
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rubén Leta
- Cardiac Imaging Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Alberico Luigi Catapano
- IRCCS Multimedica, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Giuseppe Danilo Norata
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello B, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Fernando Civeira
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Lipid Unit and Molecular Research Laboratory, IIS Aragón, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, Spain
| | - Vicenta Llorente-Cortes
- Group of Lipids and Cardiovascular Pathology, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Institute of Biomedical Research of Barcelona (IIBB) - Spanish National Research Council (CSIC), Barcelona, Spain.
| |
Collapse
|
42
|
Mauro C, Smith J, Cucchi D, Coe D, Fu H, Bonacina F, Baragetti A, Cermenati G, Caruso D, Mitro N, Catapano AL, Ammirati E, Longhi MP, Okkenhaug K, Norata GD, Marelli-Berg FM. Obesity-Induced Metabolic Stress Leads to Biased Effector Memory CD4 + T Cell Differentiation via PI3K p110δ-Akt-Mediated Signals. Cell Metab 2017; 25:593-609. [PMID: 28190771 PMCID: PMC5355363 DOI: 10.1016/j.cmet.2017.01.008] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/29/2016] [Accepted: 01/11/2017] [Indexed: 01/25/2023]
Abstract
Low-grade systemic inflammation associated to obesity leads to cardiovascular complications, caused partly by infiltration of adipose and vascular tissue by effector T cells. The signals leading to T cell differentiation and tissue infiltration during obesity are poorly understood. We tested whether saturated fatty acid-induced metabolic stress affects differentiation and trafficking patterns of CD4+ T cells. Memory CD4+ T cells primed in high-fat diet-fed donors preferentially migrated to non-lymphoid, inflammatory sites, independent of the metabolic status of the hosts. This was due to biased CD4+ T cell differentiation into CD44hi-CCR7lo-CD62Llo-CXCR3+-LFA1+ effector memory-like T cells upon priming in high-fat diet-fed animals. Similar phenotype was observed in obese subjects in a cohort of free-living people. This developmental bias was independent of any crosstalk between CD4+ T cells and dendritic cells and was mediated via direct exposure of CD4+ T cells to palmitate, leading to increased activation of a PI3K p110δ-Akt-dependent pathway upon priming.
Collapse
Affiliation(s)
- Claudio Mauro
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| | - Joanne Smith
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Danilo Cucchi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK; Istituto Pasteur, Fondazione Cenci Bolognetti, Rome 00161, Italy
| | - David Coe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Hongmei Fu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy
| | - Gaia Cermenati
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy; IRCCS Multimedica, Milan 2-242091, Italy
| | - Enrico Ammirati
- De Gasperis Cardio Center, Niguarda Ca' Granda Hospital, Milan 3-20162, Italy
| | - Maria P Longhi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan 9-20133, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Federica M Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
43
|
Yang H, Fogo AB, Kon V. Kidneys: key modulators of high-density lipoprotein levels and function. Curr Opin Nephrol Hypertens 2017; 25:174-9. [PMID: 27008596 DOI: 10.1097/mnh.0000000000000217] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The review will examine advances in our understanding of the role kidneys play in high-density lipoprotein (HDL) metabolism and the effect on levels, composition, and function of HDL particles. RECENT FINDINGS Components of the HDL particles can cross the glomerular filtration barrier. Some of these components, including apolipoproteins and enzymes involved in lipid metabolism, are taken up by the proximal tubule and degraded, modified, salvaged/returned to the circulation, or lost in the urine. Injury of the glomerular capillaries or tubules can affect these intrarenal processes and modify HDL. Changes in the plasma and urine levels of HDL may be novel markers of kidney damage or mechanism(s) of kidney disease. SUMMARY The kidneys have a significant role in the metabolism of individual HDL components, which in turn modulate HDL levels, composition, and functionality of HDL particles. These intrarenal effects may be useful markers of kidney damage and have consequences on kidney-related perturbations in HDL.
Collapse
Affiliation(s)
- Haichun Yang
- aDepartments of Pediatrics, Microbiology, and Immunology bDepartments of Medicine, Microbiology, and Immunology cDepartments of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | |
Collapse
|
44
|
Bowe B, Xie Y, Xian H, Balasubramanian S, Zayed MA, Al-Aly Z. High Density Lipoprotein Cholesterol and the Risk of All-Cause Mortality among U.S. Veterans. Clin J Am Soc Nephrol 2016; 11:1784-1793. [PMID: 27515591 PMCID: PMC5053782 DOI: 10.2215/cjn.00730116] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/08/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVES The relationship between HDL cholesterol and all-cause mortality in patients with kidney disease is not clear. We sought to characterize the relationship of HDL cholesterol and risk of death and examine the association by eGFR levels. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We built a cohort of 1,764,986 men who were United States veterans with at least one eGFR between October of 2003 and September of 2004 and followed them until September of 2013 or death. RESULTS Patients with low HDL cholesterol and low eGFR had a higher burden of comorbid illnesses. Over a median of 9.1 years (interquartile range, 7.7-9.4 years), 26,247 (40.1%), 109,222 (32.3%), 152,625 (29.2%), 113,785 (28.5%), and 139,803 (31.8%) participants with HDL cholesterol ≤25, >25 to <34, ≥34 to ≤42, >42 to <50, and ≥50 mg/dl died. In adjusted survival models, compared with the referent group of patients with low HDL cholesterol (≤25 mg/dl), intermediate HDL cholesterol levels (>25 to <34, ≥34 to ≤42, and >42 to <50 mg/dl) were associated with lower risk of death across all levels of eGFR. The lower risk was partially abrogated in those with high HDL cholesterol (≥50 mg/dl), and the risk of death was similar to the referent category among those with eGFR<30 or ≥90 ml/min per 1.73 m2. Analysis by HDL cholesterol deciles and spline analyses suggest that the relationship between HDL cholesterol and death follows a U-shaped curve. There was a significant interaction between eGFR and HDL cholesterol in that lower eGFR attenuated the salutary association of HDL cholesterol and risk of death (P for interaction <0.01). Presence of coronary artery disease attenuated the lower risk of high HDL cholesterol and all-cause mortality in those with eGFR≥60 ml/min per 1.73 m2 (P for interaction <0.05). CONCLUSIONS Our results show a U-shaped relationship between HDL cholesterol and risk of all-cause mortality across all eGFR categories. The risk is modified by eGFR and cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin Bowe
- Clinical Epidemiology Center, Research and Education Service
| | - Yan Xie
- Clinical Epidemiology Center, Research and Education Service
| | - Hong Xian
- Clinical Epidemiology Center, Research and Education Service
- Department of Biostatistics, College for Public Health and Social Justice, St. Louis University, St. Louis, Missouri; and
| | | | - Mohamed A. Zayed
- Clinical Epidemiology Center, Research and Education Service
- Section of Vascular Surgery, Surgery Service, and
- Division of Vascular and Endovascular Surgery, Department of Surgery and
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Education Service
- Division of Nephrology, Department of Medicine, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
45
|
Wang Y, Qiu X, Lv L, Wang C, Ye Z, Li S, Liu Q, Lou T, Liu X. Correlation between Serum Lipid Levels and Measured Glomerular Filtration Rate in Chinese Patients with Chronic Kidney Disease. PLoS One 2016; 11:e0163767. [PMID: 27695128 PMCID: PMC5047470 DOI: 10.1371/journal.pone.0163767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 09/13/2016] [Indexed: 01/08/2023] Open
Abstract
Introduction Dyslipidemia is often detected in patients with chronic kidney disease (CKD). Previous studies of the relationship between lipid profiles and kidney function have yielded variable results. We aimed to investigate the correlation between serum lipid levels and kidney function evaluated by measured glomerular filtration rate (mGFR) in Chinese patients with CKD. Methods A cross-sectional study was conducted on 2036 Chinese CKD patients who had mGFR. Linear regression analysis was performed to evaluate the correlation between different serum lipid levels and mGFR, while logistic regression analysis was used to investigate the association between CKD stages and the risk of different types of dyslipidemia. Results The mean age was 55 years and the mean mGFR was 63 mL/min/1.73m2. After adjusting for some confounders (age, gender, body mass index, a history of diabetes, fasting glucose, a history of hypertension, systolic blood pressure, diastolic blood pressure, smoking status, hemoglobin, serum potassium, serum albumin, and serum uric acid), serum triglyceride level showed a negative correlation with mGFR (β = -0.006, P = 0.006) in linear regression analysis, and CKD stages were positively related to the risk of hypertriglyceridemia (odds ratios were 1.329, 1.868, 2.514 and P were 0.046, < 0.001, < 0.001 for CKD stage 2, 3, 4/5, respectively) in logistic regression anlysis. Conclusions Serum triglyceride level is independently association with mGFR. Patients with reduced kidney function are more likely to have higher serum triglyceride levels. Further longitudinal, multicenter and well-conducted studies are needed to provide more evidence.
Collapse
Affiliation(s)
- Yanni Wang
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Nephrology, Hainan General Hospital, Haikou, China
| | - Xilian Qiu
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linsheng Lv
- Operation Room, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Caixia Wang
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zengchun Ye
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaomin Li
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiong Liu
- School of Software Engineerning, South China University of Tecchonology, Guangzhou, China
| | - Tanqi Lou
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- * E-mail: (XL); (TL)
| | - Xun Liu
- Department of Nephrology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- * E-mail: (XL); (TL)
| |
Collapse
|
46
|
Norata GD, Tavori H, Pirillo A, Fazio S, Catapano AL. Biology of proprotein convertase subtilisin kexin 9: beyond low-density lipoprotein cholesterol lowering. Cardiovasc Res 2016; 112:429-42. [PMID: 27496869 DOI: 10.1093/cvr/cvw194] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/06/2016] [Indexed: 12/17/2022] Open
Abstract
Proprotein convertase subtilisin kexin 9 (PCSK9) is a key regulator of low-density lipoprotein receptor levels and LDL-cholesterol levels. Loss-of-function mutations in PCSK9 gene are associated with hypocholesterolaemia and protection against cardiovascular disease, identifying PCSK9 inhibition as a valid therapeutic approach to manage hypercholesterolaemia and related diseases. Although PCSK9 is expressed mainly in the liver, it is present also in other tissues and organs with specific functions, raising the question of whether a pharmacological inhibition of PCSK9 to treat hypercholesterolaemia and associated cardiovascular diseases might be helpful or deleterious in non-hepatic tissues. For example, PCSK9 is expressed in the vascular wall, in the kidneys, and in the brain, where it was proposed to play a role in development, neurocognitive process, and neuronal apoptosis. A link between PCSK9 and immunity was also proposed as both sepsis and viral infections are differentially affected in the presence or absence of PCSK9. Despite the increasing number of observations, the debate on the exact roles of PCSK9 in extrahepatic tissues is still ongoing, and as very effective drugs that inhibit PCSK9 have become available to the clinician, a better understanding of the biological roles of PCSK9 is warranted.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy IRCCS Multimedica, Milan, Italy
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
47
|
Effect of Statins on the Progression of Coronary Calcification in Kidney Transplant Recipients. PLoS One 2016; 11:e0151797. [PMID: 27100788 PMCID: PMC4839705 DOI: 10.1371/journal.pone.0151797] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/25/2016] [Indexed: 12/26/2022] Open
Abstract
Background Coronary calcification (CAC) is highly prevalent in kidney transplant recipients (KTRs) and has been associated with cardiovascular morbidity and mortality. Some studies have shown a reduction in CAC progression with statin therapy in the general and chronic kidney disease (CKD) populations. Objectives and Methods The aim of the present study was to evaluate the effect of statins on CAC progression in incident kidney transplant recipients. Patients were randomly assigned to the statin (n = 61, 10 mg daily) and control group (n = 59). CAC and biochemical analyses were performed at baseline and 12 months. Results At baseline, CAC was observed in 30% and 21% of patients in the statin and control groups, respectively (p = 0.39). The calcium score at baseline and its absolute and relative changes over 12 months of follow up were similar among the groups. In the statin group, total cholesterol (p < 0.001), low density lipoprotein cholesterol (p < 0.001) and triglycerides (p = 0.005) decreased, and the estimated glomerular function rate increased (p<0.001) significantly. CRP levels remained stable (p = 0.52) in the statin group but increased in the control group (p = 0.01). In the multivariate model, there was no difference in CAC progression between the groups (group effect p = 0.034; time-effect p = 0.23; interaction p = 0.74). Similar results were obtained when only patients with ≥ 10AU calcium score (calcified) were analyzed (group effect p = 0.051; time-effect p = 0.58; interaction p = 0.99). Conclusion Although statins reduce the levels of cholesterol, triglycerides, inflammation and improve graft function, the dose adopted in the current study did not delay CAC progression within 12 months of follow up. Trial Registration Brazilian Clinical Trials Registry RBR-32RFMB
Collapse
|
48
|
Wahl P, Ducasa GM, Fornoni A. Systemic and renal lipids in kidney disease development and progression. Am J Physiol Renal Physiol 2016; 310:F433-45. [PMID: 26697982 PMCID: PMC4971889 DOI: 10.1152/ajprenal.00375.2015] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/22/2015] [Indexed: 12/14/2022] Open
Abstract
Altered lipid metabolism characterizes proteinuria and chronic kidney diseases. While it is thought that dyslipidemia is a consequence of kidney disease, a large body of clinical and experimental studies support that altered lipid metabolism may contribute to the pathogenesis and progression of kidney disease. In fact, accumulation of renal lipids has been observed in several conditions of genetic and nongenetic origins, linking local fat to the pathogenesis of kidney disease. Statins, which target cholesterol synthesis, have not been proven beneficial to slow the progression of chronic kidney disease. Therefore, other therapeutic strategies to reduce cholesterol accumulation in peripheral organs, such as the kidney, warrant further investigation. Recent advances in the understanding of the biology of high-density lipoprotein (HDL) have revealed that functional HDL, rather than total HDL per se, may protect from both cardiovascular and kidney diseases, strongly supporting a role for altered cholesterol efflux in the pathogenesis of kidney disease. Although the underlying pathophysiological mechanisms responsible for lipid-induced renal damage have yet to be uncovered, several studies suggest novel mechanisms by which cholesterol, free fatty acids, and sphingolipids may affect glomerular and tubular cell function. This review will focus on the clinical and experimental evidence supporting a causative role of lipids in the pathogenesis of proteinuria and kidney disease, with a primary focus on podocytes.
Collapse
Affiliation(s)
- Patricia Wahl
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| | - Gloria Michelle Ducasa
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
49
|
Adejumo OA, Okaka EI, Ojogwu LI. Lipid profile in pre-dialysis chronic kidney disease patients in southern Nigeria. Ghana Med J 2016; 50:44-9. [PMID: 27605724 PMCID: PMC4994483 DOI: 10.4314/gmj.v50i1.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Dyslipidaemia is one of the cardiovascular risk factors responsible for cardiovascular disease and rapid progression of chronic kidney disease (CKD) to end stage renal disease. Early detection and management of dyslipidaemia will reduce cardiovascular burden and retard progression of CKD. AIMS To determine the prevalence and pattern of dyslipidaemia in pre-dialysis CKD patients in a tertiary hospital in southern Nigeria. METHODS This was a case-control study that involved 105 consecutive pre-dialysis CKD patients recruited over two years and 105 age and sex matched control subjects. Data obtained from participants included demographics, body mass index, and aetiology of CKD. Blood sampling was done for the determination of creatinine and fasting serum lipids. P values < 0.05 were taken as significant. RESULTS The mean age of the CKD and control subjects were 46.98±16.81 and 47.57±15.97 years respectively with a male:female ratio of 1.7:1. The median atherogenic index of plasma (AIP), low density lipoprotein-cholesterol and triglyceride (TG) were significantly higher in the CKD patients while mean high density lipoprotein-cholesterol (HDL-C) was significantly lower in the CKD patients (p=<0.001). The overall prevalence of dyslipidaemia in the CKD patients was 60% which was significantly higher than 39% in the control (p=0.002). The prevalence of high AIP, elevated TG and reduced HDL-C increased with worsening renal function. Dyslipidaemia was commoner in female CKD patients (p=0.02) and those who were ≥ 45years (p=0.94). CONCLUSION Dyslipidaemia is common in pre-dialysis CKD especially in female and older patients. Some lipid abnormalities increased with worsening kidney function.
Collapse
Affiliation(s)
- Oluseyi A Adejumo
- Kidney Care Centre, University of Medical Sciences, Ondo, Ondo State, Nigeria
| | - Enajite I Okaka
- Department of Internal Medicine, University of Benin Teaching Hospital, Benin, Benin City, Edo State, Nigeria
| | - Louis I Ojogwu
- Department of Internal Medicine, University of Benin Teaching Hospital, Benin, Benin City, Edo State, Nigeria
| |
Collapse
|
50
|
Baragetti A, Pisano G, Bertelli C, Garlaschelli K, Grigore L, Fracanzani AL, Fargion S, Norata GD, Catapano AL. Subclinical atherosclerosis is associated with Epicardial Fat Thickness and hepatic steatosis in the general population. Nutr Metab Cardiovasc Dis 2016; 26:141-153. [PMID: 26777475 DOI: 10.1016/j.numecd.2015.10.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Abdominal obesity and hepatic steatosis are ectopic fat depots associated with Metabolic Syndrome (MetS). Epicardial Fat Thickness (EFT) is a newly discovered one, increasing with obesity, insulin resistance and MetS. Therefore we studied whether different ectopic fat markers, and EFT in particular, are associated with MetS and markers of subclinical cardiovascular disease. METHODS AND RESULTS 868 subjects from the PLIC Study were included, EFT, aortic calcifications, carotid Intima-Media Thickness (c-IMT) and echocardiographic parameters were determined by ultrasound; extra-cardiac atherosclerotic lesions were defined in presence of plaques at both carotid and aortic levels. Hepatic steatosis degrees were defined according to a scoring system. Abdominal adiposity was determined using Dual X-ray Absorbimetry (DEXA). Independently from age, women showed higher EFT versus men (4.5 (0.20-9.00) mm vs 4.00 (0.10-8.00) mm, p = 0.013); EFT was thicker in post-menopausal women (independently from hormone-replacement therapy). EFT, liver steatosis and abdominal adiposity increased with MetS (p < 0.001). EFT was the only ectopic fat marker associated with cardiac dysfunction (OR = 1.340 [1.088-1.651 95% C.I., p = 0.006); liver steatosis and EFT were associated with extra-cardiac plaques (OR = 2.529 [1.328-4.819] 95% C.I., p < 0.001 and OR = 1.195 [1.008-1.299] 95% C.I., p = 0.042; respectively). On top of cardiovascular risk factors, only EFT improved the discrimination of subjects with cardiac dysfunction and atherosclerotic plaques. CONCLUSIONS EFT is associated with left ventricular dysfunction and subclinical atherosclerosis. Our data suggest that EFT may represent an additional tool for the stratification of cardiovascular risk.
Collapse
Affiliation(s)
- A Baragetti
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - G Pisano
- Dipartimento di Medicina Interna, Centro Studi Malattie Metaboliche del Fegato - Cà Granda IRCCS Fondazione Ospedale Policlinico, Milan, Italy
| | - C Bertelli
- Dipartimento di Medicina Interna, Centro Studi Malattie Metaboliche del Fegato - Cà Granda IRCCS Fondazione Ospedale Policlinico, Milan, Italy
| | - K Garlaschelli
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - L Grigore
- Center for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - A L Fracanzani
- Dipartimento di Medicina Interna, Centro Studi Malattie Metaboliche del Fegato - Cà Granda IRCCS Fondazione Ospedale Policlinico, Milan, Italy
| | - S Fargion
- Dipartimento di Medicina Interna, Centro Studi Malattie Metaboliche del Fegato - Cà Granda IRCCS Fondazione Ospedale Policlinico, Milan, Italy
| | - G D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; William Harvey Research Institute, Barts and The London School of Medicine & Dentistry Queen's Mary University, London, United Kingdom.
| | - A L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; Multimedica Hospital - IRCCS, Sesto San Giovanni, Milan, Italy.
| |
Collapse
|