1
|
Su J, Fan X, Zou Y, Fu G, Feng S, Wang X, Yu Y, Li L, Bian Z, Huang R, Qin L, Chen J, Zeng Q, Yan K, Gao C, Lian Z, Li X, Li Y. Inhibition of Aberrant Activated Fibroblast-Like Synoviocytes in Rheumatoid Arthritis by Leishmania Peptide via the Regulation of Fatty Acid Synthesis Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409154. [PMID: 40125636 PMCID: PMC12097062 DOI: 10.1002/advs.202409154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 03/06/2025] [Indexed: 03/25/2025]
Abstract
The Leishmania homolog of receptors for activated C kinase (LACK) protein is derived from Leishmania parasites L. major. The polypeptide LACK156-173 has been shown to confer protection against murine autoimmune arthritis. Fibroblast-like synoviocytes (FLSs) play a pivotal role in the synovial invasion and joint destruction observed in rheumatoid arthritis (RA). The study reveals that LACK156-173 can inhibit the aggressive phenotype of RA-FLSs by restoring dysregulated fatty acid synthesis metabolism. In RA-FLSs, overexpression of fatty acid synthase (FASN) leads to excessive fatty acid accumulation, which in turn promotes mitochondrial fragmentation by enhancing phosphorylation at the ser616 site of dynamin 1-like protein (DRP1). This process increases reactive oxygen species (ROS) production and activates the PI3K/mTOR/NF-κB pathway, thereby facilitating the transition of RA-FLSs to an aggressive inflammatory and bone-damaging phenotype. LACK156-173 is internalized into the cytoplasm via CAPN2-mediated endocytosis, where it directly binds to FASN and inhibits its activity. The findings suggest that targeting the restoration of fatty acid metabolism could potentially alleviate synovial invasion and joint damage in RA. LACK156-173 may therefore hold therapeutic promise for RA patients.
Collapse
Affiliation(s)
- Jianling Su
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510000China
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Xuemei Fan
- Department of RheumatologyZibo Central HospitalZiboShandong255036China
| | - Yaoyao Zou
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Guangtao Fu
- Department of OrthopedicsGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Shiqi Feng
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510000China
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Xiaoxue Wang
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhen UniversityShenzhen518035China
| | - Yongmei Yu
- Department of Rheumatology and Immunology2nd Affiliated Hospital of Harbin Medical UniversityHarbin150001China
| | - Lin Li
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510000China
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Zhenhua Bian
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhou511442China
| | - Rongrong Huang
- Department of PharmacyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Linmang Qin
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Jiping Chen
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Qin Zeng
- Guangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510000China
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Kai Yan
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Caiyue Gao
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhexiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| | - Xin Li
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Yang Li
- Department of Rheumatology and ImmunologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000China
| |
Collapse
|
2
|
Zhang X, Hu H, He L, Huang X, Zhang Z, Tu L, Zhang H, Liu F, Liu X, He Y. Association between triglyceride to high-density lipoprotein cholesterol ratio and microalbuminuria in the Chinese population. Sci Rep 2024; 14:30960. [PMID: 39730606 DOI: 10.1038/s41598-024-82084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024] Open
Abstract
The evidence for the association between the triglyceride (TG) to high-density lipoprotein cholesterol (HDL-c) ratio and the risk of developing microalbuminuria is still limited in the Chinese population. Therefore, our research will endeavor to explore the relationship between the two. The cross-sectional survey enrolled 32,877 general population from eight regional centers in China. If the urinary albumin-creatinine ratio (UACR) ≥ 30 mg/g, it was considered microalbuminuria. The dependent variable in the study was microalbuminuria, while the independent variable was the TG/HDL-c ratio. A binary logistic regression model was utilized to examine the independent association between the likelihood of microalbuminuria and the TG/HDL-c ratio. A generalized additive model (GAM) and spline smoothing were employed to investigate non-linear relationships between these two variables. To confirm the findings and pinpoint important turning points, subgroup and threshold effect tests were performed. The average age of the study participants was 57.67 ± 9.29 years, with 22,052 (67%) females and 10,825 (33%) males. The ratio of TG/HDL-c had a median value of 1.06, while the UACR had a median value of 9.92 mg/g. The prevalence of microalbuminuria was 14.4%. Following covariate adjustment, the prevalence of microalbuminuria and the TG/HDL-c ratio showed a significant positive connection (OR = 1.17,95% CI 1.13-1.21). Interestingly, we discovered a non-linear relationship between the occurrence of microalbuminuria and the TG/HDL-c ratio. At a TG/HDL-c ratio of 0.911, we detected an inflection point, with ORs of 1.12 (95% CI: 1.08, 1.17) on the right side of the point and 1.89 (95% CI: 1.51, 2.36) on the left. Subgroup analyses further validated the results. Our research revealed a positive and non-linear connection between the TG/HDL-c ratio and the prevalence of microalbumin in Chinese individuals. The connection was especially robust when the TG/HDL-c ratio was below 0.911. Reducing TG or raising HDL-c levels in order to lower the TG/HDL-c ratio may significantly mitigate the risk of microalbuminuria.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China
| | - Haofei Hu
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, Guangdong Province, China
| | - Lishu He
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Xia Huang
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China
| | - Zhichao Zhang
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China
| | - Lirong Tu
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China
| | - Heping Zhang
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China
| | - Feiyuan Liu
- Department of Nephrology, Langzhong Hospital of T.C.M, No.333 Badu Avenue, Langzhong, Nanchong, 637400, Sichuan Province, China
| | - Xiaohui Liu
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China.
| | - Yongcheng He
- Department of Nephrology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Rd, Nanchong, 637000, Sichuan Province, China.
| |
Collapse
|
3
|
Yang Q, Zou Y, Lang Y, Yang J, Wu Y, Xiao X, Qin C, Zhao Y, Liu F. Estimated small dense low-density lipoprotein-cholesterol and the risk of kidney and cardiovascular outcomes in diabetic kidney disease. Ren Fail 2024; 46:2369701. [PMID: 38952279 PMCID: PMC467091 DOI: 10.1080/0886022x.2024.2369701] [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: 07/31/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
AIMS This study aimed to investigate the correlations between estimated small dense low-density lipoprotein-cholesterol (esd-LDL-c) and the development of end-stage kidney disease (ESKD), cardiovascular mortality, and all-cause mortality in individuals with diabetic kidney disease (DKD) or diabetes mellitus (DM) concomitant chronic kidney disease (CKD). METHODS We analyzed the data from a biopsy-proven DKD cohort conducted at West China Hospital of Sichuan University between 2009 and 2021 (the DKD cohort) and participants with DM and CKD in the National Health and Nutrition Examination Survey (NHANES) 2011-2014 (the NHANES DM-CKD cohort). Cox regression analysis was also used to estimate associations between esd-LDL-c and the incidence of ESKD, cardiovascular mortality, and all-cause mortality. RESULTS There were 175 ESKD events among 338 participants in the DKD cohort. Patients were divided into three groups based on esd-LDL-c tertiles (T1 < 33.7 mg/dL, T2 ≥ 33.7 mg/dL to <45.9 mg/dL, T3 ≥ 45.9 mg/dL). The highest tertile of esd-LDL-c was associated with ESKD (adjusted HR 2.016, 95% CI 1.144-3.554, p = .015). Furthermore, there were 99 deaths (39 cardiovascular) among 293 participants in the NHANES DM-CKD cohort. Participants were classified into three groups in line with the tertile values of esd-LDL-c in the DKD cohort. The highest tertile of esd-LDL-c was associated with cardiovascular mortality (adjusted HR 3.95, 95% CI 1.3-12, p = .016) and all-cause mortality (adjusted HR 2.37, 95% CI 1.06-5.32, p = .036). CONCLUSIONS Higher esd-LDL-c was associated with increased risk of ESKD in people with biopsy-proven DKD, and higher cardiovascular and all-cause mortality risk among those with DM-CKD.
Collapse
Affiliation(s)
- Qing Yang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Yutong Zou
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Yanlin Lang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Jia Yang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Yucheng Wu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Xiang Xiao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Chunmei Qin
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Yuancheng Zhao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Bomholt AB, Johansen CD, Galsgaard KD, Elmelund E, Winther-Sørensen M, Holst JJ, Wewer Albrechtsen NJ, Sørensen CM. Glucagon receptor activation contributes to the development of kidney injury. Am J Physiol Renal Physiol 2024; 327:F712-F724. [PMID: 39265079 PMCID: PMC11563637 DOI: 10.1152/ajprenal.00088.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
The underlying causes of diabetic kidney disease are still largely unknown. New insights into the contributing causes of diabetic nephropathy are important to prevent this complication. Hyperglycemia and hypertension are some of the risk factors for diabetic nephropathy. However, the incidence of diabetic nephropathy is increasing despite efforts to normalize blood glucose levels and blood pressure. Therefore, other factors should be investigated as causes of diabetic nephropathy. We investigated whether long-term increased plasma levels of glucagon contribute to the development of pathophysiological changes in kidney function as seen in patients with diabetic nephropathy. Using mouse models of chronic activation and inactivation of glucagon receptor signaling, we investigated whether glucagon is involved in changes in renal function, renal structure, and transcriptional changes. We found several histopathological changes in the kidney, such as thickening of the parietal layer of Bowman's capsule, glomerular mesangial cell expansion, and significant albuminuria in the mice with activated glucagon receptor signaling. Opposite effects on mesangial area expansion and the development of albuminuria were demonstrated in mice with glucagon receptor inactivation. RNA sequencing data revealed that transcription of genes related to fatty acid metabolism, podocytes, Na+-K+-ATPase, and sodium/glucose transport was significantly changed in mice with activated glucagon receptor signaling. These data implicate that glucagon receptor signaling is involved in the development of kidney injury, as seen in type 2 diabetes, and that glucagon receptor is a potential therapeutic target in the treatment of diabetes. NEW & NOTEWORTHY This study suggests that the glucagon receptor is a potential therapeutic target in the treatment of diabetic kidney disease. We show, in mice, that long-term treatment with a glucagon analog showed not only pathophysiological changes and changes in renal function but also transcriptional changes in the kidneys, whereas opposite effects were demonstrated in mice with glucagon receptor inactivation. Therefore, the use of glucagon in a treatment regimen requires investigation of possible metabolic and renal abnormalities.
Collapse
Affiliation(s)
| | - Christian Dall Johansen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Elmelund
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital-Bispebjerg Hospital, Copenhagen, Denmark
| | | |
Collapse
|
5
|
Pan J, Li C, Zhang J, Sun Z, Yu X, Wan Q, Ruan Z, Wang W, Li Y. Association between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) and diabetic kidney disease in patients with diabetes in the United States: a cross-sectional study. Lipids Health Dis 2024; 23:317. [PMID: 39334123 PMCID: PMC11437633 DOI: 10.1186/s12944-024-02308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND This paper investigated the link between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) and diabetic kidney disease (DKD) in adult diabetic patients and identified the optimal NHHR value for impacting DKD. METHODS This cross-sectional research made use of records from the National Health and Nutrition Examination Survey (NHANES) executed between 2005 and 2016. The link of NHHR to DKD risk was analyzed by logistic regression and restricted cubic spline (RCS) models. The stability and reliability of the results were assessed by subgroup analysis and sensitivity analysis. RESULTS In total, 4,177 participants were involved. As a continuous variable, NHHR was markedly connected to an increased risk of DKD (OR 1.07, 95% CI 1.02, 1.12, P < 0.01). When NHHR was grouped in quartiles, relative to the reference set, the highest NHHR group was also linked to a heightened risk of DKD (OR 1.23, 95% CI 1.01, 1.50, P < 0.05). The outcome of RCS show a "J" shaped correlation between NHHR and DKD risk (P for nonlinear = 0.0136). The risk of developing DKD was the lowest when NHHR equals 2.66. Subgroup analysis revealed that the link of NHHR to DKD persisted in participants aged below 40, females, non-smokers, and those without hyperuricemia. Sensitivity analysis demonstrated a certain robustness in this association. CONCLUSION A meaningful link is present between NHHR and DKD. An NHHR value of around 2.66 could represent the ideal cutoff for assessing DKD risk.
Collapse
Affiliation(s)
- Jingjing Pan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Changnian Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Jiayi Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Zhenhua Sun
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Xiaoying Yu
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Qianhui Wan
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Zhishen Ruan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China
| | - Wenbo Wang
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China.
- Department of Orthopaedics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, NO.1 Jingba Road, Jinan, 250000, Shandong Province, People's Republic of China.
| | - Yujie Li
- Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People's Republic of China.
- Department of Geriatrics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, NO.1 Jingba Road, Jinan, 250000, Shandong Province, People's Republic of China.
| |
Collapse
|
6
|
Chen Y, Chen M, Zhu W, Zhang Y, Liu P, Li P. Morroniside attenuates podocytes lipid deposition in diabetic nephropathy: A network pharmacology, molecular docking and experimental validation study. Int Immunopharmacol 2024; 138:112560. [PMID: 38959541 DOI: 10.1016/j.intimp.2024.112560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Dysregulation of lipid metabolism is a key factor influencing the progression of diabetic nephropathy (DN). Morroniside (MOR) is a major active compound isolated from the traditional Chinese herb Cornus officinalis, our previous research found that it can improve the lipid deposition of renal tubular epithelial cells. The purpose of this study is to explore whether MOR can improve podocyte lipid deposition and its mechanism of reducing DN. METHODS Initially, we used network pharmacology and bioinformatics techniques to predict the relationship between renal lipid metabolism of MOR and DN. Subsequently, the binding activity of MOR with lipid-related proteins was studied by molecular docking to determine how MOR acts through these proteins. After determining the target of MOR, animal experiments and cell tests were carried out to verify it. RESULTS Using network pharmacology, bioinformatics, and molecular docking, target proteins for MOR treatment of DN were predicted and screened, including PGC-1α, LXRs, ABCA1, PPARY, CD36, and nephrin. It is particularly noted that MOR effectively binds to PGC-1α, while LXRs, ABCA1, PPARY and CD36 are downstream molecules of PGC-1α. Silencing the PGC-1α gene significantly reduced the therapeutic effects of MOR. Conversely, in groups without PGC-1α knockdown, MOR was able to increase the expression levels of PGC-1α and influence the expression of downstream proteins. Furthermore, through in vivo and in vitro experiments, utilizing techniques such as lipid droplet staining, PAS, MASSON staining, immunofluorescence, and Western blot, we found that MOR effectively elevated the expression levels of the podocyte protein nephrin and lipid metabolism-regulating proteins PGC-1α, PPARY, and ABCA1, while significantly inhibiting the expression of the lipid accumulation promoter CD36. CONCLUSION MOR can regulate the cholesterol efflux in podocytes via the PGC-1α/LXRs/ABCA1 signaling pathway, and control cholesterol intake via the PGC-1α/PPARY/CD36 signaling pathway, thereby ameliorating lipid deposition in DN.
Collapse
Affiliation(s)
- Yao Chen
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Ming Chen
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Wenhui Zhu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yonggang Zhang
- First People's Hospital of Qiqihaer City, Heilongjiang Province, China
| | - Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China.
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
7
|
Mao TH, Huang HQ, Zhang CH. Clinical characteristics and treatment compounds of obesity-related kidney injury. World J Diabetes 2024; 15:1091-1110. [PMID: 38983811 PMCID: PMC11229974 DOI: 10.4239/wjd.v15.i6.1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/22/2023] [Accepted: 04/08/2024] [Indexed: 06/11/2024] Open
Abstract
Disorders in energy homeostasis can lead to various metabolic diseases, particularly obesity. The obesity epidemic has led to an increased incidence of obesity-related nephropathy (ORN), a distinct entity characterized by proteinuria, glomerulomegaly, progressive glomerulosclerosis, and renal function decline. Obesity and its associated renal damage are common in clinical practice, and their incidence is increasing and attracting great attention. There is a great need to identify safe and effective therapeutic modalities, and therapeutics using chemical compounds and natural products are receiving increasing attention. However, the summary is lacking about the specific effects and mechanisms of action of compounds in the treatment of ORN. In this review, we summarize the important clinical features and compound treatment strategies for obesity and obesity-induced kidney injury. We also summarize the pathologic and clinical features of ORN as well as its pathogenesis and potential therapeutics targeting renal inflammation, oxidative stress, insulin resistance, fibrosis, kidney lipid accumulation, and dysregulated autophagy. In addition, detailed information on natural and synthetic compounds used for the treatment of obesity-related kidney disease is summarized. The synthesis of detailed information aims to contribute to a deeper understanding of the clinical treatment modalities for obesity-related kidney diseases, fostering the anticipation of novel insights in this domain.
Collapse
Affiliation(s)
- Tuo-Hua Mao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Han-Qi Huang
- Department of Endocrinology, Hubei No. 3 People’s Hospital of Jianghan University, Wuhan 430033, Hubei Province, China
| | - Chuan-Hai Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
8
|
Han YZ, Du BX, Zhu XY, Wang YZY, Zheng HJ, Liu WJ. Lipid metabolism disorder in diabetic kidney disease. Front Endocrinol (Lausanne) 2024; 15:1336402. [PMID: 38742197 PMCID: PMC11089115 DOI: 10.3389/fendo.2024.1336402] [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: 11/10/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic kidney disease (DKD), a significant complication associated with diabetes mellitus, presents limited treatment options. The progression of DKD is marked by substantial lipid disturbances, including alterations in triglycerides, cholesterol, sphingolipids, phospholipids, lipid droplets, and bile acids (BAs). Altered lipid metabolism serves as a crucial pathogenic mechanism in DKD, potentially intertwined with cellular ferroptosis, lipophagy, lipid metabolism reprogramming, and immune modulation of gut microbiota (thus impacting the liver-kidney axis). The elucidation of these mechanisms opens new potential therapeutic pathways for DKD management. This research explores the link between lipid metabolism disruptions and DKD onset.
Collapse
Affiliation(s)
- Yi-Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing-Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang-Zhi-Yuan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei-Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Li X, Bhattacharya D, Yuan Y, Wei C, Zhong F, Ding F, D'Agati VD, Lee K, Friedman SL, He JC. Chronic kidney disease in a murine model of non-alcoholic steatohepatitis (NASH). Kidney Int 2024; 105:540-561. [PMID: 38159678 PMCID: PMC10922588 DOI: 10.1016/j.kint.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024]
Abstract
Clinical studies suggest that non-alcoholic steatohepatitis (NASH) is an independent risk factor for chronic kidney disease (CKD), but causality and mechanisms linking these two major diseases are lacking. To assess whether NASH can induce CKD, we have characterized kidney function, histological features, transcriptomic and lipidomic profiles in a well-validated murine NASH model. Mice with NASH progressively developed significant podocyte foot process effacement, proteinuria, glomerulosclerosis, tubular epithelial cell injury, lipid accumulation, and interstitial fibrosis. The progression of kidney fibrosis paralleled the severity of the histologic NASH-activity score. Significantly, we confirmed the causal link between NASH and CKD by orthotopic liver transplantation, which attenuated proteinuria, kidney dysfunction, and fibrosis compared with control sham operated mice. Transcriptomic analysis of mouse kidney cortices revealed differentially expressed genes that were highly enriched in mitochondrial dysfunction, lipid metabolic process, and insulin signaling pathways in NASH-induced CKD. Lipidomic analysis of kidney cortices further revealed that phospholipids and sphingolipids were the most significantly changed lipid species. Notably, we found similar kidney histological changes in human NASH and CKD. Thus, our results confirm a causative role of NASH in the development of CKD, reveal potential pathophysiologic mechanisms of NASH-induced kidney injury, and established a valuable model to study the pathogenesis of NASH-associated CKD. This is an important feature of fatty liver disease that has been largely overlooked but has clinical and prognostic importance.
Collapse
Affiliation(s)
- Xuezhu Li
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yue Yuan
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Chengguo Wei
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fang Zhong
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Feng Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Vivette D D'Agati
- Department of Pathology, Columbia University Medical Center, New York, New York, USA
| | - Kyung Lee
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - John Cijiang He
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Renal Program, James J Peters VA Medical Center at Bronx, New York, New York, USA.
| |
Collapse
|
10
|
He M, Wei W, Zhang Y, Xiang Z, Peng D, Kasimumali A, Rong S. Gut microbial metabolites SCFAs and chronic kidney disease. J Transl Med 2024; 22:172. [PMID: 38369469 PMCID: PMC10874542 DOI: 10.1186/s12967-024-04974-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024] Open
Abstract
The global incidence of Chronic Kidney Disease (CKD) is steadily escalating, with discernible linkage to the intricate terrain of intestinal microecology. The intestinal microbiota orchestrates a dynamic equilibrium in the organism, metabolizing dietary-derived compounds, a process which profoundly impacts human health. Among these compounds, short-chain fatty acids (SCFAs), which result from microbial metabolic processes, play a versatile role in influencing host energy homeostasis, immune function, and intermicrobial signaling, etc. SCFAs emerge as pivotal risk factors influencing CKD's development and prognosis. This paper review elucidates the impact of gut microbial metabolites, specifically SCFAs, on CKD, highlighting their role in modulating host inflammatory responses, oxidative stress, cellular autophagy, the immune milieu, and signaling cascades. An in-depth comprehension of the interplay between SCFAs and kidney disease pathogenesis may pave the way for their utilization as biomarkers for CKD progression and prognosis or as novel adjunctive therapeutic strategies.
Collapse
Affiliation(s)
- Meng He
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenqian Wei
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhouxia Xiang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dan Peng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ayijiaken Kasimumali
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
11
|
Luo Q, Wei Y, Lv X, Chen W, Yang D, Tuo Q. The Effect and Mechanism of Oleanolic Acid in the Treatment of Metabolic Syndrome and Related Cardiovascular Diseases. Molecules 2024; 29:758. [PMID: 38398510 PMCID: PMC10892503 DOI: 10.3390/molecules29040758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Metabolic syndromes (MetS) and related cardiovascular diseases (CVDs) pose a serious threat to human health. MetS are metabolic disorders characterized by obesity, dyslipidemia, and hypertension, which increase the risk of CVDs' initiation and development. Although there are many availabile drugs for treating MetS and related CVDs, some side effects also occur. Considering the low-level side effects, many natural products have been tried to treat MetS and CVDs. A five-cyclic triterpenoid natural product, oleanolic acid (OA), has been reported to have many pharmacologic actions such as anti-hypertension, anti-hyperlipidemia, and liver protection. OA has specific advantages in the treatment of MetS and CVDs. OA achieves therapeutic effects through a variety of pathways, attracting great interest and playing a vital role in the treatment of MetS and CVDs. Consequently, in this article, we aim to review the pharmacological actions and potential mechanisms of OA in treating MetS and related CVDs.
Collapse
Affiliation(s)
- Quanye Luo
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Yu Wei
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Xuzhen Lv
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, The School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China;
| | - Wen Chen
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Dongmei Yang
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| | - Qinhui Tuo
- Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.L.); (Y.W.); (W.C.)
| |
Collapse
|
12
|
Liu J, Pan S, Wang X, Liu Z, Zhang Y. Role of advanced glycation end products in diabetic vascular injury: molecular mechanisms and therapeutic perspectives. Eur J Med Res 2023; 28:553. [PMID: 38042909 PMCID: PMC10693038 DOI: 10.1186/s40001-023-01431-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/04/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND In diabetic metabolic disorders, advanced glycation end products (AGEs) contribute significantly to the development of cardiovascular diseases (CVD). AIMS This comprehensive review aims to elucidate the molecular mechanisms underlying AGE-mediated vascular injury. CONCLUSIONS We discuss the formation and accumulation of AGEs, their interactions with cellular receptors, and the subsequent activation of signaling pathways leading to oxidative stress, inflammation, endothelial dysfunction, smooth muscle cell proliferation, extracellular matrix remodeling, and impaired angiogenesis. Moreover, we explore potential therapeutic strategies targeting AGEs and related pathways for CVD prevention and treatment in diabetic metabolic disorders. Finally, we address current challenges and future directions in the field, emphasizing the importance of understanding the molecular links between AGEs and vascular injury to improve patient outcomes.
Collapse
Affiliation(s)
- Jing Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
| | - Shuo Pan
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
| | - Xiqiang Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
| | - Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
- Affiliated Shaanxi Provincial People's Hospital, Medical Research Institute, Northwestern Polytechnical University, Xi'an, China.
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
| |
Collapse
|
13
|
Sawie HG, Khadrawy YA, El-Gizawy MM, Mourad HH, Omara EA, Hosny EN. Effect of alpha-lipoic acid and caffeine-loaded chitosan nanoparticles on obesity and its complications in liver and kidney in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3017-3031. [PMID: 37306714 PMCID: PMC10567965 DOI: 10.1007/s00210-023-02507-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/19/2023] [Indexed: 06/13/2023]
Abstract
The present work investigated the effect of α-lipoic acid (ALA) and caffeine-loaded chitosan nanoparticles (CAF-CS NPs) on obesity and its hepatic and renal complications in rats. Rats were divided into control, rat model of obesity induced by high fat diet (HFD), and obese rats treated with ALA and/or CAF-CS NPs. At the end of the experiment, the activities of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) and the levels of urea, creatinine, interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) were determined in the sera of animals. In addition, malondialdehyde (MDA), nitric oxide (NO), and reduced glutathione (GSH) were measured in hepatic and renal tissues. Renal Na+, K+-ATPase was assessed. The histopathological changes were examined in the hepatic and renal tissues. Obese rats showed a significant increase in AST, ALT, ALP, urea, and creatinine. This was associated with a significant increase in IL-1β, TNF-α, MDA, and NO. A significant decrease in hepatic and renal GSH and renal Na+, K+-ATPase activity was recorded in obese rats. Obese rats also showed histopathological alterations in hepatic and renal tissues. Treatment with ALA and/or CAF-CS NPs reduced the weight of obese rats and ameliorated almost all the hepatic and renal biochemical and histopathological changes induced in obese rats. In conclusion, the present findings indicate that ALA and/or CAF-CS NPs offered an effective therapy against obesity induced by HFD and its hepatic and renal complications. The therapeutic effect of ALA and CAF-CS NPs could be mediated through their antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Hussein G Sawie
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt
| | - Mayada M El-Gizawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt
| | - Hagar H Mourad
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt
| | - Enayat A Omara
- Pathology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Eman N Hosny
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt.
| |
Collapse
|
14
|
Yang D, Yang X, Chen S, Lv M, Tan J, Yang D. Ox-LDL aggravates contrast-induced injury of renal tubular epithelial cells. J Biochem Mol Toxicol 2023; 37:e23379. [PMID: 37186061 DOI: 10.1002/jbt.23379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/04/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023]
Abstract
Hypercholesterolemia can aggravate contrast-induced acute kidney injury, and the exacerbation of renal tubular epithelial cell (RTEC) injury is a major cause. However, the exact mechanisms remain obscure. Mitophagy, a type of autophagy, selectively eliminates damaged mitochondria and reduces mitochondrial oxidative stress, which is strongly implicated in cell homeostasis and acute kidney injury. Oxidized low-density lipoprotein (Ox-LDL) is accumulated in hypercholesterolemia and has a cytotoxic effect. This study aimed to determine whether and how ox-LDL exacerbates contrast-induced injury in RTECs and to further explore whether PINK1/Parkin-dependent mitophagy is involved in this process. Iohexol and ox-LDL were used alone or in combination to treat HK-2 cells. Rapamycin pretreatment was utilized to enhance mitophagy. Cell viability, apoptosis, mitochondrial membrane potential (MMP) and mitochondrial reactive oxygen species (mtROS) were detected by cell counting kit-8, TUNEL staining, JC-1 kit and MitoSOX fluorescence, respectively. The expression of mitophagy-related proteins (including PINK1, Parkin, and so on) and cleaved caspase-3 was confirmed by western blot. Colocalization of MitoTracker-labeled mitochondria and LysoTracker-labeled lysosomes was observed by fluorescence microscopy to evaluate mitophagy. The results of our study showed that ox-LDL aggravated MMP decline, mtROS release and apoptosis in iohexol-treated HK-2 cells, accompanied by a further increased autophagy level. Enhancement of PINK1/Parkin-dependent mitophagy by rapamycin alleviated apoptosis and mitochondrial injury in HK-2 cells in response to iohexol under ox-LDL condition. Therefore, our findings indicate that ox-LDL aggravates contrast-induced injury of RTECs by increasing mitochondrial damage and mitochondrial oxidative stress, which may be associated with the relative insufficiency of PINK1/Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Dingwei Yang
- Department of Nephrology, Tianjin Hospital of Tianjin University, Tianjin, China
| | - Xueyan Yang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sha Chen
- Department of Nephrology, Tianjin Hospital of Tianjin University, Tianjin, China
| | - Meiling Lv
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Dingping Yang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
15
|
Chen X, Ran J, Mazhar M, Zhu Y, Lin Y, Qin L, Miao S. The balanced unsaturated fatty acid supplement constituted by woody edible oils improved lipid metabolism and gut microbiota in high-fat diet mice. Front Nutr 2023; 10:1203932. [PMID: 37545586 PMCID: PMC10399753 DOI: 10.3389/fnut.2023.1203932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
The dietary intervention has demonstrated effectiveness in improving hyperlipidemia and obesity. Woody edible oils are rich in unsaturated fatty acids (UFAs) that could positively affect lipid metabolism. In this study, the blended oil (BLO), a balanced UFA supplement, constituted by Zanthoxylum bungeanum (Chinese Red Pepper) seed oil, walnut (Juglans regia) oil, camellia (Camema oleifera) seed oil and perilla (Perilla frutescens) seed oil was established referring to the Chinese dietary reference intakes, in which the ratios of monounsaturated/polyunsaturated fatty acids and ω-6/ω-3 polyunsaturated fatty acids were 1:1 and 4:1, respectively. The BLO was administrated to KM mice fed a high-fat diet (HFD) by gavage every day at a dose of 3.0 mL/kg·bw for 10 weeks to assess its effects on serum lipid levels, liver antioxidant activities and gut microbial composition. The results showed that the BLO improved hepatic steatosis, liver oxidative stress, and serum lipid levels. Additionally, there was an increased abundance of Lactobacillus, Allobaculum, and Blautia, along with a decreased abundance of Staphylococcus in cecal contents. These changes were found to be positively correlated with the metabolic improvements, as indicated by Spearman's correlation analysis. These findings implied the practicality of the balanced unsaturated fatty acid consumption in preventing hyperlipidemia and obesity.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- Department of Laboratory Medicine, Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, China
| | - Jingqi Ran
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Muhammad Mazhar
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Yong Zhu
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Yichen Lin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Likang Qin
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Song Miao
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| |
Collapse
|
16
|
Shnayder NA, Grechkina VV, Trefilova VV, Efremov IS, Dontceva EA, Narodova EA, Petrova MM, Soloveva IA, Tepnadze LE, Reznichenko PA, Al-Zamil M, Altynbekova GI, Strelnik AI, Nasyrova RF. Valproate-Induced Metabolic Syndrome. Biomedicines 2023; 11:biomedicines11051499. [PMID: 37239168 DOI: 10.3390/biomedicines11051499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Valproic acid (VPA) and its salts (sodium calcium magnesium and orotic) are psychotropic drugs that are widely used in neurology and psychiatry. The long-term use of VPA increases the risk of developing adverse drug reactions (ADRs), among which metabolic syndrome (MetS) plays a special role. MetS belongs to a cluster of metabolic conditions such as abdominal obesity, high blood pressure, high blood glucose, high serum triglycerides, and low serum high-density lipoprotein. Valproate-induced MetS (VPA-MetS) is a common ADR that needs an updated multidisciplinary approach to its prevention and diagnosis. In this review, we consider the results of studies of blood (serum and plasma) and the urinary biomarkers of VPA-MetS. These metabolic biomarkers may provide the key to the development of a new multidisciplinary personalized strategy for the prevention and diagnosis of VPA-MetS in patients with neurological diseases, psychiatric disorders, and addiction diseases.
Collapse
Affiliation(s)
- Natalia A Shnayder
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Violetta V Grechkina
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
| | - Vera V Trefilova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Department of Neurology, Hospital for War Veterans, 193079 Saint Petersburg, Russia
| | - Ilya S Efremov
- Department of Psychiatry and Narcology, Bashkir State Medical University, 450008 Ufa, Russia
| | - Evgenia A Dontceva
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Federal Centre for Neurosurgery, 630087 Novosibirsk, Russia
| | - Ekaterina A Narodova
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Marina M Petrova
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Irina A Soloveva
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Liia E Tepnadze
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Polina A Reznichenko
- Shared Core Facilities "Molecular and Cell Technologies", V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples' Friendship University of Russia, 117198 Moscow, Russia
| | - Gulnara I Altynbekova
- Department of Psychiatry and Narcology, S.D. Asfendiarov Kazakh National Medical University, Almaty 050022, Kazakhstan
| | - Anna I Strelnik
- International Centre for Education and Research in Neuropsychiatry, Samara State Medical University, 443016 Samara, Russia
| | - Regina F Nasyrova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- International Centre for Education and Research in Neuropsychiatry, Samara State Medical University, 443016 Samara, Russia
| |
Collapse
|
17
|
Tikhonova I, Dyukina A, Shaykhutdinova E, Safronova V. Modified Signaling of Membrane Formyl Peptide Receptors in NADPH-Oxidase Regulation in Obesity-Resistant Mice. MEMBRANES 2023; 13:306. [PMID: 36984693 PMCID: PMC10058262 DOI: 10.3390/membranes13030306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
The signaling of membrane receptors is modified in obesity characterized by low-grade inflammation. The obesity-resistant state of organisms is poorly understood. We analyzed the generation of reactive oxygen species (ROS) initiated though membrane formyl peptide receptors (Fpr1, Fpr2) in bone-marrow granulocytes of obesity-resistant mice (ORM). A chemiluminescence assay was used to assess NADPH-oxidase-related intensity of ROS generation. ORM were chosen from animals that received high-fat diets and had metric body parameters as controls (standard diet). High spontaneous ROS production was observed in ORM cells. The EC50 for responses to bacterial or mitochondrial peptide N-formyl-MLF was higher in ORM with and without inflammation vs. the same control groups, indicating an insignificant role of high-affinity Fpr1. Increased responses to synthetic peptide WKYMVM (Fpr2 agonist) were observed in controls with acute inflammation, but they were similar in other groups. Fpr2 was possibly partially inactivated in ORM owing to the inflammatory state. Weakened Fpr1 and Fpr2 signaling via MAPKs was revealed in ORM using specific inhibitors for p38, ERK1/2, and JNK. P38 signaling via Fpr2 was lower in ORM with inflammation. Thus, a high-fat diet modified FPRs' role and suppressed MAPK signaling in NADPH-oxidase regulation in ORM. This result can be useful to understand the immunological features of obesity resistance.
Collapse
Affiliation(s)
- Irina Tikhonova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya St., 3, 142290 Pushchino, Russia
| | - Alsu Dyukina
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya St., 3, 142290 Pushchino, Russia
| | - Elvira Shaykhutdinova
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospect Nauki, 6, 142290 Pushchino, Russia
| | - Valentina Safronova
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya St., 3, 142290 Pushchino, Russia
| |
Collapse
|
18
|
Chen X, Shi C, Wang Y, Yu H, Zhang Y, Zhang J, Li P, Gao J. The mechanisms of glycolipid metabolism disorder on vascular injury in type 2 diabetes. Front Physiol 2022; 13:952445. [PMID: 36117707 PMCID: PMC9473659 DOI: 10.3389/fphys.2022.952445] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with diabetes have severe vascular complications, such as diabetic nephropathy, diabetic retinopathy, cardiovascular disease, and neuropathy. Devastating vascular complications lead to increased mortality, blindness, kidney failure, and decreased overall quality of life in people with type 2 diabetes (T2D). Glycolipid metabolism disorder plays a vital role in the vascular complications of T2D. However, the specific mechanism of action remains to be elucidated. In T2D patients, vascular damage begins to develop before insulin resistance and clinical diagnosis. Endothelial dysregulation is a significant cause of vascular complications and the early event of vascular injury. Hyperglycemia and hyperlipidemia can trigger inflammation and oxidative stress, which impair endothelial function. Furthermore, during the pathogenesis of T2D, epigenetic modifications are aberrant and activate various biological processes, resulting in endothelial dysregulation. In the present review, we provide an overview and discussion of the roles of hyperglycemia- and hyperlipidemia-induced endothelial dysfunction, inflammatory response, oxidative stress, and epigenetic modification in the pathogenesis of T2D. Understanding the connections of glucotoxicity and lipotoxicity with vascular injury may reveal a novel potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Xiatian Chen
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Yin Wang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hua Yu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Yu Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiaxuan Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Yang Z, Zhu X, Wen A, Ran J, Qin L, Zhu Y. Coix Seed-Based Milk Fermented With Limosilactobacillus reuteri Improves Lipid Metabolism and Gut Microbiota in Mice Fed With a High-Fat Diet. Front Nutr 2022; 9:921255. [PMID: 35903451 PMCID: PMC9320324 DOI: 10.3389/fnut.2022.921255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to investigate the effects of coix seed-based milk (CSM) fermented with Limosilactobacillus reuteri (L. reuteri) on dyslipidemia and the composition of the intestinal microbiota in high fat diet (HFD)-fed mice. Changes in the body weight, serum lipid levels, activities of hepatic oxidative stress factors, expression of lipid-related genes, and composition of the intestinal microbiota of HFD-fed mice after supplementation with CSM were determined. The results showed that intake of CSM reduced the body weight gain as well as serum total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C) levels, and increased the high-density lipoprotein cholesterol (HDL-C) levels in the mice. Meanwhile, supplementation with CSM could relieve liver oxidative stress, down-regulate the expression of genes related to lipid synthesis, and prevent liver fat accumulation in mice fed with HFD. The 16S rRNA sequencing of the intestinal microbiota showed that CSM regulated the gut microbiota community structure at different taxonomic levels, and reversed gut dysbiosis induced by HFD. The relative abundance of Muribaculaceae, Lachnospiraceae, Dubosiella and Akkermansia which are negatively correlated with blood lipid levels were significantly increased by the intervention of CSM, while the relative abundance of Desulfovibrionaceae, Ruminococca-ceae_UCG-014, Psychrobacter, and Staphylococcus which have positive correlation with blood lipid levels were significantly decreased. These results indicated that CSM might serve as a novel and promising dietary supplement for ameliorating hyperlipidemia and intestinal microbiota disorders caused by HFDs.
Collapse
Affiliation(s)
- Zhoujie Yang
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
| | - Xiaoli Zhu
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Anyan Wen
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Jingqi Ran
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Likang Qin
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
- *Correspondence: Likang Qin
| | - Yi Zhu
- Plant Protection and Plant Quarantine Station of Guizhou Province, Guiyang, China
| |
Collapse
|
20
|
Wang Y, Fan Z, Yang M, Wang Y, Cao J, Khan A, Liu Y, Cheng G. Protective effects of E Se tea extracts against alcoholic fatty liver disease induced by high fat/alcohol diet: In vivo biological evaluation and molecular docking study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154113. [PMID: 35490493 DOI: 10.1016/j.phymed.2022.154113] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND With the development of economy and increased workload, chronic a high-fat/alcohol diet intake may lead to alcoholic fatty liver disease (AFLD), which is considered as a crucial health problem worldwide. E Se tea is produced of the leaves and leaf buds of Malus toringoides (Rehd.) Hughes in Tibet and has human health benefits with anti-hyperglycemia, hypertension, and hyperlipidemia effects. PURPOSE The objective of this work was to investigate the protective effect of aqueous-ethanol and hot-water extracts of E Se tea against chronic high-fat/alcohol diet induced AFLD rats. METHODS Firstly, to determine the chemical profiling of E Se tea extracts, UHPLC-ESI-HRMS analysis was conducted. Secondly, Sprague-Dawley male rats were used to establish the AFLD animal model by feeding with high-fat/alcohol diet. The animals were treated with E Se tea extracts for 12 weeks. Serum parameters were determined, histologic sections were prepared, and activities of enzymes related to inflammatory response and lipid metabolism imbalance were analyzed. The underlying mechanisms of E Se tea extracts alleviating AFLD were analyzed by immunofluorescence staining and Western blotting analysis. Lastly, key targets of 11-MT against AFLD were verified through molecular docking. RESULTS In this study, seven main compounds were confirmed or tentatively identified in E Se tea extracts by UHPLC-ESI-HRMS. The results revealed that both the extracts could reverse histopathological steatotic alternation of the liver and reduced the activity of liver damage markers (ALT, AST). E Se tea extracts mitigated oxidative stress by inhibiting CYP2E1 protein and lipid peroxidation parameters (MDA), but enhancing the endogenous antioxidants (CAT, GSH, SOD). Moreover, E Se tea extracts ameliorated inflammation by restraining the activation of NF-κB, consequently releasing the expression of proinflammatory cytokines (TNF-α, IL-6, IL-1β, COX-2 and iNOS). Subsequently, E Se tea extracts reduced hepatocyte apoptosis by increasing capase-9, caspase-3 and Bax protein expression but decreasing Bcl-2 protein expression. Furthermore, E Se tea extracts improved metabolism imbalance by stimulating AMPK/SREBP1/FAS and PPAR-α/CPT1 signaling pathway by regulating lipid metabolism parameters (TC, TG, HDL-C, LHD-C). Furthermore, molecular docking results indicated that 7 chemical constituents of E Se tea extracts had strong docking affinity with 4 key target proteins (AMPK, PPAR-α, NF-кB and Caspase-9). CONCLUSION E Se tea ameliorated AFLD through ameliorating inflammatory response, apoptosis, and lipid metabolism imbalance.
Collapse
Affiliation(s)
- Yongpeng Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhifeng Fan
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; College of Ethnic Medicine, Yunnan Minzu University, Kunming, 650500, China
| | - Meilian Yang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yudan Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; College of Ethnic Medicine, Yunnan Minzu University, Kunming, 650500, China; National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, Yunnan Minzu University, Kunming, 650500, China
| | - Jianxin Cao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| | - Yaping Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Guiguang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
21
|
Camastra S, Ferrannini E. Role of anatomical location, cellular phenotype and perfusion of adipose tissue in intermediary metabolism: A narrative review. Rev Endocr Metab Disord 2022; 23:43-50. [PMID: 35031911 PMCID: PMC8873050 DOI: 10.1007/s11154-021-09708-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 02/07/2023]
Abstract
It is well-established that adipose tissue accumulation is associated with insulin resistance through multiple mechanisms. One major metabolic link is the classical Randle cycle: enhanced release of free fatty acids (FFA) from hydrolysis of adipose tissue triglycerides impedes insulin-mediated glucose uptake in muscle tissues. Less well studied are the different routes of this communication. First, white adipose tissue depots may be regionally distant from muscle (i.e., gluteal fat and diaphragm muscle) or contiguous to muscle but separated by a fascia (Scarpa's fascia in the abdomen, fascia lata in the thigh). In this case, released FFA outflow through the venous drainage and merge into arterial plasma to be transported to muscle tissues. Next, cytosolic triglycerides can directly, i.e., within the cell, provide FFA to myocytes (but also pancreatic ß-cells, renal tubular cells, etc.). Finally, adipocyte layers or lumps may be adjacent to, but not anatomically segregated, from muscle, as is typically the case for epicardial fat and cardiomyocytes. As regulation of these three main delivery paths is different, their separate contribution to substrate competition at the whole-body level is uncertain. Another important link between fat and muscle is vascular. In the resting state, blood flow is generally higher in adipose tissue than in muscle. In the insulinized state, fat blood flow is directly related to whole-body insulin resistance whereas muscle blood flow is not; consequently, fractional (i.e., flow-adjusted) glucose uptake is stimulated in muscle but not fat. Thus, reduced blood supply is a major factor for the impairment of in vivo insulin-mediated glucose uptake in both subcutaneous and visceral fat. In contrast, the insulin resistance of glucose uptake in resting skeletal muscle is predominantly a cellular defect.
Collapse
Affiliation(s)
- Stefania Camastra
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy.
| | | |
Collapse
|
22
|
Ertuglu L, Yildiz A, Gamboa J, Ikizler TA. Skeletal muscle energetics in patients with moderate to advanced kidney disease. Kidney Res Clin Pract 2022; 41:14-21. [PMID: 35108768 PMCID: PMC8816417 DOI: 10.23876/j.krcp.21.175] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Sarcopenia, defined as decrease in muscle function and mass, is common in patients with moderate to advanced chronic kidney disease (CKD) and is associated with poor clinical outcomes. Muscle mitochondrial dysfunction is proposed as one of the mechanisms underlying sarcopenia. Patients with moderate to advanced CKD have decreased muscle mitochondrial content and oxidative capacity along with suppressed activity of various mitochondrial enzymes such as mitochondrial electron transport chain complexes and pyruvate dehydrogenase, leading to impaired energy production. Other mitochondrial abnormalities found in this population include defective beta-oxidation of fatty acids and mitochondrial DNA mutations. These changes are noticeable from the early stages of CKD and correlate with severity of the disease. Damage induced by uremic toxins, oxidative stress, and systemic inflammation has been implicated in the development of mitochondrial dysfunction in CKD patients. Given that mitochondrial function is an important determinant of physical activity and performance, its modulation is a potential therapeutic target for sarcopenia in patients with kidney disease. Coenzyme Q, nicotinamide, and cardiolipin-targeted peptides have been tested as therapeutic interventions in early studies. Aerobic exercise, a well-established strategy to improve muscle function and mass in healthy adults, is not as effective in patients with advanced kidney disease. This might be due to reduced expression or impaired activation of peroxisome proliferator-activated receptor-gamma coactivator 1α, the master regulator of mitochondrial biogenesis. Further studies are needed to broaden our understanding of the pathogenesis of mitochondrial dysfunction and to develop mitochondrial-targeted therapies for prevention and treatment of sarcopenia in patients with CKD.
Collapse
Affiliation(s)
- Lale Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abdulmecit Yildiz
- Division of Nephrology, Department of Medicine, Uludag University, Bursa, Turkey
| | - Jorge Gamboa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - T. Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Health Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Correspondence: T. Alp Ikizler Division of Nephrology, Vanderbilt University Medical Center, 1161 21st Avenue South, S-3223 Medical Center North, Nashville 37232, TN, USA. E-mail:
| |
Collapse
|
23
|
Yan Q, Zhao Z, Liu D, Li J, Pan S, Duan J, Dong J, Liu Z. Integrated analysis of potential gene crosstalk between non-alcoholic fatty liver disease and diabetic nephropathy. Front Endocrinol (Lausanne) 2022; 13:1032814. [PMID: 36387855 PMCID: PMC9642911 DOI: 10.3389/fendo.2022.1032814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Growing evidence indicates that non-alcoholic fatty liver disease (NAFLD) is related to the occurrence and development of diabetic nephropathy (DN). This bioinformatics study aimed to explore optimal crosstalk genes and related pathways between NAFLD and DN. METHODS Gene expression profiles were downloaded from Gene Expression Omnibus. CIBERSORT algorithm was employed to analyze the similarity of infiltrating immunocytes between the two diseases. Protein-protein interaction (PPI) co-expression network and functional enrichment analysis were conducted based on the identification of common differentially expressed genes (DEGs). Least absolute shrinkage and selection operator (LASSO) regression and Boruta algorithm were implemented to initially screen crosstalk genes. Machine learning models, including support vector machine, random forest model, and generalized linear model, were utilized to further identify the optimal crosstalk genes between DN and NAFLD. An integrated network containing crosstalk genes, transcription factors, and associated pathways was developed. RESULTS Four gene expression datasets, including GSE66676 and GSE48452 for NAFLD and GSE30122 and GSE1009 for DN, were involved in this study. There were 80 common DEGs between the two diseases in total. The PPI network built with the 80 common genes included 77 nodes and 83 edges. Ten optimal crosstalk genes were selected by LASSO regression and Boruta algorithm, including CD36, WIPI1, CBX7, FCN1, SLC35D2, CP, ZDHHC3, PTPN3, LPL, and SPP1. Among these genes, LPL and SPP1 were the most significant according to NAFLD-transcription factor network. Five hundred twenty-nine nodes and 1,113 edges comprised the PPI network of activated pathway-gene. In addition, 14 common pathways of these two diseases were recognized using Gene Ontology (GO) analysis; among them, regulation of the lipid metabolic process is closely related to both two diseases. CONCLUSIONS This study offers hints that NAFLD and DN have a common pathogenesis, and LPL and SPP1 are the most relevant crosstalk genes. Based on the common pathways and optimal crosstalk genes, our proposal carried out further research to disclose the etiology and pathology between the two diseases.
Collapse
Affiliation(s)
- Qianqian Yan
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
| | - Zihao Zhao
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
| | - Dongwei Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jia Li
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jiayu Duan
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- *Correspondence: Jiayu Duan, ; Jiancheng Dong, ; Zhangsuo Liu,
| | - Jiancheng Dong
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jiayu Duan, ; Jiancheng Dong, ; Zhangsuo Liu,
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- *Correspondence: Jiayu Duan, ; Jiancheng Dong, ; Zhangsuo Liu,
| |
Collapse
|
24
|
Lim ZW, Chen WL. Polymorphism rs10105606 of LPL as a Novel Risk Factor for Microalbuminuria. J Inflamm Res 2021; 14:6833-6844. [PMID: 34934334 PMCID: PMC8684407 DOI: 10.2147/jir.s338010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction An important clinical feature of metabolic syndrome is abdominal obesity. Microalbuminuria is important in predicting the risk of cardiovascular and renal complications in abdominal obesity patients. However, the association between microalbuminuria polymorphism and abdominal obesity has not been conducted. The objective of this study is to analyze the genetic polymorphism of microalbuminuria in participants with metabolically unhealthy obesity (MUO). Methods Among 1325 MUO participants, we identified genomic loci underlying those with microalbuminuria, compared to those without microalbuminuria. Single nucleotide polymorphisms (SNPs) were selected with P < 1 × 10−5 from the Manhattan plot. Multivariable linear regression and analysis of variance were used to analyze the association between different SNP genotypes and microalbuminuria. Results The analysis showed homozygous participants for the risk allele A of rs10105606 and Affx-31885823 had 1.978-fold risk and 1.921-fold increased risk of microalbuminuria, respectively. Heterozygous distribution of rs117180252, rs10105606, and Affx-31885823 also increased the risk of microalbuminuria compared to the wild type. Further analysis showed Lipoprotein lipase (LPL), RN7SL87P, and RPL30P9 were the candidate genes associated with lipid metabolism and abdominal obesity. Conclusion In conclusion, LPL, RN7SL87P, and RPL30P9 minor allele carriers with abdominal obesity are more susceptible to microalbuminuria, explaining the inter-individual differences of microalbuminuria in MUO patients.
Collapse
Affiliation(s)
- Zhu Wei Lim
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan, Republic of China
| | - Wei Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
25
|
Wu Z, Ma Q, Cai S, Sun Y, Zhang Y, Yi J. Rhus chinensis Mill. Fruits Ameliorate Hepatic Glycolipid Metabolism Disorder in Rats Induced by High Fat/High Sugar Diet. Nutrients 2021; 13:nu13124480. [PMID: 34960032 PMCID: PMC8708379 DOI: 10.3390/nu13124480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatic glycolipid metabolism disorder is considered as one of the key factors in the pathogenesis of many chronic diseases. The objective of this study was to investigate the protective effect and underlying mechanisms of Rhus chinensis Mill. fruits against hepatic glycolipid metabolic disorders in rats induced by a high fat/high sugar diet. Results showed that ethanol extract, especially at a dose of 600 mg/kg b.w., could effectively ameliorate glycolipid metabolic disorders in rats. The biochemical indexes, including CAT, GSH and HOMA-IR, were significantly improved by the administration of ethanol extract. Immunohistochemistry and Western blot analysis revealed that ethanol extract up-regulated the expression levels of PI3K/AKT, PPAR-α, and the phosphorylation of IRS1 and AMPK proteins, and down-regulated the expressions of SREBP-1 and FAS proteins in the liver, which are closely related to hepatic glycolipid metabolism. Those findings suggested that R. chinensis Mill. fruits could be developed as functional foods and/or nutraceuticals for preventing or controlling some chronic diseases related to hepatic glycolipid metabolism disorder.
Collapse
Affiliation(s)
- Zihuan Wu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (Z.W.); (S.C.); (Y.S.); (Y.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Qingqing Ma
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (Z.W.); (S.C.); (Y.S.); (Y.Z.)
| | - Yilin Sun
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (Z.W.); (S.C.); (Y.S.); (Y.Z.)
| | - Yuanyue Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (Z.W.); (S.C.); (Y.S.); (Y.Z.)
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (Z.W.); (S.C.); (Y.S.); (Y.Z.)
- Correspondence: ; Tel.: +86-15810687441
| |
Collapse
|
26
|
Oxidized LDL Is Associated with eGFR Decline in Proteinuric Diabetic Kidney Disease: A Cohort Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2968869. [PMID: 34712380 PMCID: PMC8548137 DOI: 10.1155/2021/2968869] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/28/2021] [Accepted: 09/14/2021] [Indexed: 11/26/2022]
Abstract
Diabetic kidney disease (DKD) is a highly heterogenous disease, including the proteinuric and the nonproteinuric pattern. Oxidized low-density lipoprotein (ox-LDL) is progressively increased in DKD and causes direct damage to kidney tubular epithelial cells through a mechanism similar to that underlying the deleterious effect of lipid peroxides in the vascular endothelium. We aimed to examine the association between plasma ox-LDL cholesterol and clinical endpoints in DKD patients. Ninety-one patients with established proteinuric DKD and diabetic retinopathy were enrolled and prospectively followed for 10 years or the occurrence of death, or at least 30% decline in eGFR, or progression to end-stage kidney disease (ESKD) requiring renal replacement therapy (primary outcome). At the end of the study, both eGFR and proteinuria were reassessed. Secondary outcomes of the study were the percentage change in eGFR and proteinuria over time for each patient. At baseline, patients were divided into 2 groups according to the median ox-LDL value (i.e., below or equal and above 66.22 U/L). Both Kaplan-Meier curves (p = 0.001, log-rank test) and univariate Cox regression analysis showed that high ox-LDL was associated with the primary outcome (HR = 3.42, 95%CI = 1.55 − 7.56, p = 0.002). After adjustment for various well-known cofounders, multivariate Cox analysis showed that the association between increased circulating ox-LDL levels and the composite kidney endpoint remained significant (HR = 2.87, 95%CI = 1.14–7.20, p = 0.025). Regarding the secondary outcome of eGFR decline, the assessment of areas under the curves (AUC) showed that ox-LDL outperformed several cofounding factors (AUC 71%, 95%CI = 0.59 − 0.83, p = 0.001) and had better accuracy to predict deterioration of eGFR over time than baseline proteinuria (AUC 67%, 95%CI = 0.54 − 0.79, p = 0.014). Increased ox-LDL might be associated with disease progression in proteinuric DKD.
Collapse
|
27
|
Ossoli A, Strazzella A, Rottoli D, Zanchi C, Locatelli M, Zoja C, Simonelli S, Veglia F, Barbaras R, Tupin C, Dasseux JL, Calabresi L. CER-001 ameliorates lipid profile and kidney disease in a mouse model of familial LCAT deficiency. Metabolism 2021; 116:154464. [PMID: 33309714 DOI: 10.1016/j.metabol.2020.154464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/25/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE CER-001 is an HDL mimetic that has been tested in different pathological conditions, but never with LCAT deficiency. This study was designed to investigate whether the absence of LCAT affects the catabolic fate of CER-001, and to evaluate the effects of CER-001 on kidney disease associated with LCAT deficiency. METHODS Lcat-/- and wild-type mice received CER-001 (2.5, 5, 10 mg/kg) intravenously for 2 weeks. The plasma lipid/ lipoprotein profile and HDL subclasses were analyzed. In a second set of experiments, Lcat-/- mice were injected with LpX to induce renal disease and treated with CER-001 and then the plasma lipid profile, lipid accumulation in the kidney, albuminuria and glomerular podocyte markers were evaluated. RESULTS In Lcat-/- mice a decrease in total cholesterol and triglycerides, and an increase in HDL-c was observed after CER-001 treatment. While in wild-type mice CER-001 entered the classical HDL remodeling pathway, in the absence of LCAT it disappeared from the plasma shortly after injection and ended up in the kidney. In a mouse model of renal disease in LCAT deficiency, treatment with CER-001 at 10 mg/kg for one month had beneficial effects not only on the lipid profile, but also on renal disease, by limiting albuminuria and podocyte dysfunction. CONCLUSIONS Treatment with CER-001 ameliorates the dyslipidemia typically associated with LCAT deficiency and more importantly limits renal damage in a mouse model of renal disease in LCAT deficiency. The present results provide a rationale for using CER-001 in FLD patients.
Collapse
Affiliation(s)
- Alice Ossoli
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Arianna Strazzella
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Daniela Rottoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Cristina Zanchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Monica Locatelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Sara Simonelli
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | | | | | | | | | - Laura Calabresi
- Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
28
|
Zhao Y, Goto M, Vaziri ND, Khazaeli M, Liu H, Farahanchi N, Khanifar E, Farzaneh T, Haslett PA, Moradi H, Soundarapandian MM. RNA Interference Targeting Liver Angiopoietin-Like Protein 3 Protects from Nephrotic Syndrome in a Rat Model Via Amelioration of Pathologic Hypertriglyceridemia. J Pharmacol Exp Ther 2021; 376:428-435. [PMID: 33443084 DOI: 10.1124/jpet.120.000257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/04/2020] [Indexed: 11/22/2022] Open
Abstract
Nephrotic syndrome (NS) is associated with metabolic perturbances including profound dyslipidemia characterized by hypercholesterolemia and hypertriglyceridemia. A major underlying mechanism of hypertriglyceridemia in NS is lipoprotein lipase (LPL) deficiency and dysfunction. There is emerging evidence that elevated angiopoietin-like protein 3 (ANGPTL3), an LPL inhibitor that is primarily expressed and secreted by hepatocytes, may be in part responsible for these findings. Furthermore, there is evidence pointing to the contribution of ANGPTL3 to the pathogenesis of proteinuria in NS. Therefore, we hypothesized that inhibition of hepatic ANGPTL3 by RNA interference will ameliorate dyslipidemia and other symptoms of NS and pave the way for a new therapeutic strategy. To this end, we used a subcutaneously delivered, GalNAc (N-Acetylgalactosamine)-conjugated small interfering RNA (siRNA) to selectively target and suppress liver Angptl3 in rats with puromycin-induced NS, which exhibits clinical features of NS including proteinuria, hypoalbuminemia, hyperlipidemia, and renal histologic abnormalities. The study demonstrated that siRNA-mediated knockdown of the liver Angptl3 relieved its inhibitory effect on LPL and significantly reduced hypertriglyceridemia in nephrotic rats. This was accompanied by diminished proteinuria and hypoalbuminemia, which are the hallmarks of NS, and significant attenuation of renal tissue inflammation and oxidative stress. Taken together, this study confirmed the hypothesis that suppression of Angptl3 is protective in NS and points to the possibility that the use of RNA interference to suppress hepatic Angptl3 can serve as a novel therapeutic strategy for NS. SIGNIFICANCE STATEMENT: The current standard of care for mitigating nephrotic dyslipidemia in nephrotic syndrome is statins therapy. However, the efficacy of statins and its safety in the context of impaired kidney function is not well established. Here, we present an alternate therapeutic approach by using siRNA targeting Angptl3 expressed in hepatocytes. As the liver is the major source of circulating Angptl3, siRNA treatment reduced the profound hypertriglyceridemia in a rat model of nephrotic syndrome and was also effective in improving kidney and cardiac function.
Collapse
Affiliation(s)
- Yitong Zhao
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Masaki Goto
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Mahyar Khazaeli
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Han Liu
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Nazli Farahanchi
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Elham Khanifar
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Ted Farzaneh
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Patrick A Haslett
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Hamid Moradi
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| | - Mangala M Soundarapandian
- Division of Nephrology and Hypertension, University of California, Irvine, California (Y.Z., M.G., N.D.V., M.K., H.L., N.F., H.M.); Long Beach Memorial Pathology Group, Long Beach, California (E.K.); Department of Pathology and Laboratory Medicine, University of California, Irvine, California (T.F.); Tibor Rubin VA Medical Center, Department of Medicine, Nephrology Section, Long Beach, California (H.M.); and Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts (P.A.H., M.M.S.)
| |
Collapse
|
29
|
Xue J, Wang Y, Li B, Yu S, Wang A, Wang W, Gao Z, Tang X, Yan L, Wan Q, Qin G, Chen L, Ning G, Mu Y. Triglycerides to high-density lipoprotein cholesterol ratio is superior to triglycerides and other lipid ratios as an indicator of increased urinary albumin-to-creatinine ratio in the general population of China: a cross-sectional study. Lipids Health Dis 2021; 20:13. [PMID: 33588849 PMCID: PMC7883433 DOI: 10.1186/s12944-021-01442-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/02/2021] [Indexed: 11/13/2022] Open
Abstract
Background Dyslipidemia contributes to the pathogenesis of renal dysfunction. Previous research demonstrated that triglycerides (TG), instead of other individual lipid indexes, has a significant link with elevated urinary albumin-to-creatinine ratio (UACR). However, it is unclear whether lipid ratios are superior indicators of increased UACR compared with TG. This research is to determine whether there are close relationships of lipid ratios with UACR in a general population. Methods 35,751 participants from seven centers across China were enrolled. UACR equal or higher than 30 mg/g was recognized as increased albuminuria. The associations of TG, low-density lipoprotein cholesterol (LDL-C)/ high-density lipoprotein cholesterol (HDL-C), TG/HDL-C and non-high-density lipoprotein cholesterol (non-HDL-C)/HDL-C with increased UACR were evaluated by linear and logistic regression analyses in females and males separately. Results There were 3692 (14.8%) female subjects, and 1307 (12.0%) male subjects characterized as having increased UACR. There were significantly differences in TG/HDL-C and non-HDL-C/HDL-C between the normal UACR group and the increased UACR group, while LDL-C/HDL-C was not. Furthermore, linear regression analysis was implemented and showed that TG and TG/HDL-C were both positively related to UACR even after a variety of potential confounders were adjusted regardless of sexes, while the correlation between non-HDL-C/HDL-C and elevated UACR were only significant in females. Further analyses utilizing logistic regression demonstrated that compared with non-HDL-C/HDL-C and TG, TG/HDL-C showed the strongest association with increased UACR (quartile 1 of TG/HDL-C as a reference; OR [95% CI] of quartile 4: 1.28 [1.13–1.44] in women, 1.24 [1.02–1.50] in men) after fully adjusting for potential confounding factors. Stratified analyses revealed that in males who were overweight and in females who were overweight or over 55 years or had prediabetes or prehypertension, TG/HDL-C had significant associations with abnormal UACR. Conclusions Compared with TG and other routine lipid ratios, TG/HDL-C is a superior indicator for increased UACR. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01442-8.
Collapse
Affiliation(s)
- Jing Xue
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China
| | - Yuxia Wang
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China
| | - Bing Li
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China
| | - Songyan Yu
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China
| | - Anping Wang
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China
| | - Weiqing Wang
- Shanghai Jiaotong University Affiliated Ruijin Hospital, Shanghai, China
| | - Zhengnan Gao
- Center Hospital of Dalian, Dalian, Liaoning, China
| | - Xulei Tang
- First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Li Yan
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin Wan
- Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan, China
| | - Guijun Qin
- Zhengzhou University First affiliated Hospital, Zhengzhou, Henan, China
| | - Lulu Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guang Ning
- Shanghai Jiaotong University Affiliated Ruijin Hospital, Shanghai, China
| | - Yiming Mu
- Medical School of Chinese PLA, Beijing, China. .,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, No.28 Fuxing Road, Haidian district, Beijing, 100853, China.
| |
Collapse
|
30
|
Nakamichi R, Hayashi K, Itoh H. Effects of High Glucose and Lipotoxicity on Diabetic Podocytes. Nutrients 2021; 13:nu13010241. [PMID: 33467659 PMCID: PMC7830342 DOI: 10.3390/nu13010241] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/11/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Glomerular podocytes are highly differentiated cells that cover glomerular capillaries from the outside and have a characteristic morphology with numerous foot processes. The formation of slit membranes between the foot processes serves as a final filtration barrier for urine filtration from the blood. Podocyte damage causes disruption of the slit membrane, subsequent proteinuria and finally glomerulosclerosis, which is a common pathway in various types of chronic kidney disease (CKD). In recent years, there has been an increase in diabetes, due to rapid lifestyle changes, which is the main cause of CKD. Therefore, understanding the effect of diabetic status on podocytes is of great importance to establish a strategy for preventing CKD progression. In this review, we summarize altered glucose and lipid metabolism in diabetic podocytes and also discuss the reversibility of the changes in podocyte phenotype.
Collapse
Affiliation(s)
| | - Kaori Hayashi
- Correspondence: ; Tel.: +81-3-5363-3796; Fax: +81-3-3359-2745
| | | |
Collapse
|
31
|
Lv XC, Chen M, Huang ZR, Guo WL, Ai LZ, Bai WD, Yu XD, Liu YL, Rao PF, Ni L. Potential mechanisms underlying the ameliorative effect of Lactobacillus paracasei FZU103 on the lipid metabolism in hyperlipidemic mice fed a high-fat diet. Food Res Int 2021; 139:109956. [DOI: 10.1016/j.foodres.2020.109956] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022]
|
32
|
Lin PH, Duann P. Dyslipidemia in Kidney Disorders: Perspectives on Mitochondria Homeostasis and Therapeutic Opportunities. Front Physiol 2020; 11:1050. [PMID: 33013450 PMCID: PMC7494972 DOI: 10.3389/fphys.2020.01050] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
To excrete body nitrogen waste and regulate electrolyte and fluid balance, the kidney has developed into an energy factory with only second to the heart in mitochondrial content in the body to meet the high-energy demand and regulate homeostasis. Energy supply from the renal mitochondria majorly depends on lipid metabolism, with programed enzyme systems in fatty acid β-oxidation and Krebs cycle. Renal mitochondria integrate several metabolic pathways, including AMPK/PGC-1α, PPARs, and CD36 signaling to maintain energy homeostasis for dynamic and static requirements. The pathobiology of several kidney disorders, including diabetic nephropathy, acute and chronic kidney injuries, has been primarily linked to impaired mitochondrial bioenergetics. Such homeostatic disruption in turn stimulates a pathological adaptation, with mitochondrial enzyme system reprograming possibly leading to dyslipidemia. However, this alteration, while rescuing oncotic pressure deficit secondary to albuminuria and dissipating edematous disorder, also imposes an ominous lipotoxic consequence. Reprograming of lipid metabolism in kidney injury is essential to preserve the integrity of kidney mitochondria, thereby preventing massive collateral damage including excessive autophagy and chronic inflammation. Here, we review dyslipidemia in kidney disorders and the most recent advances on targeting mitochondrial energy metabolism as a therapeutic strategy to restrict renal lipotoxicity, achieve salutary anti-edematous effects, and restore mitochondrial homeostasis.
Collapse
Affiliation(s)
- Pei-Hui Lin
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Pu Duann
- Research and Development, Salem Veteran Affairs Medical Center, Salem, VA, United States
| |
Collapse
|
33
|
Liu X, Ducasa GM, Mallela SK, Kim JJ, Molina J, Mitrofanova A, Wilbon SS, Ge M, Fontanella A, Pedigo C, Santos JV, Nelson RG, Drexler Y, Contreras G, Al-Ali H, Merscher S, Fornoni A. Sterol-O-acyltransferase-1 has a role in kidney disease associated with diabetes and Alport syndrome. Kidney Int 2020; 98:1275-1285. [PMID: 32739420 DOI: 10.1016/j.kint.2020.06.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/25/2022]
Abstract
Defective cholesterol metabolism primarily linked to reduced ATP-binding cassette transporter A1 (ABCA1) expression is closely associated with the pathogenesis and progression of kidney diseases, including diabetic kidney disease and Alport Syndrome. However, whether the accumulation of free or esterified cholesterol contributes to progression in kidney disease remains unclear. Here, we demonstrate that inhibition of sterol-O-acyltransferase-1 (SOAT1), the enzyme at the endoplasmic reticulum that converts free cholesterol to cholesterol esters, which are then stored in lipid droplets, effectively reduced cholesterol ester and lipid droplet formation in human podocytes. Furthermore, we found that inhibition of SOAT1 in podocytes reduced lipotoxicity-mediated podocyte injury in diabetic kidney disease and Alport Syndrome in association with increased ABCA1 expression and ABCA1-mediated cholesterol efflux. In vivo, Soat1 deficient mice did not develop albuminuria or mesangial expansion at 10-12 months of age. However, Soat1 deficiency/inhibition in experimental models of diabetic kidney disease and Alport Syndrome reduced cholesterol ester content in kidney cortices and protected from disease progression. Thus, targeting SOAT1-mediated cholesterol metabolism may represent a new therapeutic strategy to treat kidney disease in patients with diabetic kidney disease and Alport Syndrome, like that suggested for Alzheimer's disease and cancer treatments.
Collapse
Affiliation(s)
- Xiaochen Liu
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Gloria Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shamroop Kumar Mallela
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sydney Symone Wilbon
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Antonio Fontanella
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Christopher Pedigo
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Javier Varona Santos
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Yelena Drexler
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Gabriel Contreras
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Hassan Al-Ali
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
34
|
Michael OS, Dibia CL, Adeyanju OA, Olaniyi KS, Areola ED, Olatunji LA. Estrogen-progestin oral contraceptive and nicotine exposure synergistically confers cardio-renoprotection in female Wistar rats. Biomed Pharmacother 2020; 129:110387. [PMID: 32540646 DOI: 10.1016/j.biopha.2020.110387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 10/24/2022] Open
Abstract
Approximately fifty percent of premenopausal women who smoke cigarettes or on nicotine replacement therapy are also on hormonal contraceptives, especially oral estrogen-progestin. Oral estrogen-progestin therapy has been reported to promote insulin resistance (IR) which causes lipid influx into non-adipose tissue and impairs Na+/K+ -ATPase activity, especially in the heart and kidney. However, the effects of nicotine on excess lipid and altered Na+/K+ -ATPase activity associated with the use of estrogen-progestin therapy have not been fully elucidated. This study therefore aimed at investigating the effect of nicotine on cardiac and renal lipid influx and Na+/K+ -ATPase activity during estrogen-progestin therapy. Twenty-four female Wistar rats grouped into 4 (n = 6/group) received (p.o.) vehicle, nicotine (1.0 mg/kg) with or without estrogen-progestin steroids (1.0 μg ethinyl estradiol and 5.0 μg levonorgestrel) and estrogen-progestin only daily for 6 weeks. Data showed that estrogen-progestin treatment or nicotine exposure caused IR, hyperinsulinemia, increased cardiac and renal uric acid, malondialdehyde, triglyceride, glycogen synthase kinase-3, plasminogen activator inhibitor-1, reduced bilirubin and circulating estradiol. Estrogen-progestin treatment led to decreased cardiac Na+/K+-ATPase activity while nicotine did not alter Na+/K+-ATPase activity but increased plasma and tissue cotinine. Renal Na+/K+-ATPase activity was not altered by the treatments. However, all these alterations were reversed following combined administration of oral estrogen-progestin therapy and nicotine. The present study therefore demonstrates that oral estrogen-progestin therapy and nicotine exposure synergistically prevents IR-linked cardio-renotoxicity with corresponding improvement in cardiac and renal lipid accumulation, oxidative stress, inflammation and Na+/K+-ATPase activity.
Collapse
Affiliation(s)
- O S Michael
- Cardiometabolic Research Unit, Department of Physiology, College of Health Sciences, Bowen University, Iwo, Nigeria; HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria.
| | - C L Dibia
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria; Department of Physiology, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - O A Adeyanju
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, 360101, Nigeria
| | - K S Olaniyi
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, 360101, Nigeria
| | - E D Areola
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - L A Olatunji
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
35
|
Pessoa EDA, Convento MB, Castino B, Leme AM, de Oliveira AS, Aragão A, Fernandes SM, Carbonel A, Dezoti C, Vattimo MDF, Schor N, Borges FT. Beneficial Effects of Isoflavones in the Kidney of Obese Rats Are Mediated by PPAR-Gamma Expression. Nutrients 2020; 12:nu12061624. [PMID: 32492810 PMCID: PMC7352183 DOI: 10.3390/nu12061624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Several studies have demonstrated an important association between altered lipid metabolism and the development of kidney injury because of a high-fat diet. Fructose is also closely associated with renal injury. We opted for a combination of fructose and saturated fats in a diet (DH) that is a model known to induce renal damage in order to evaluate whether soy isoflavones could have promising use in the treatment of renal alterations. After two months of ingestion, there was an expansion of visceral fat, which was associated with long-term metabolic disorders, such as sustained hyperglycemia, insulin resistance, polyuria, dyslipidemia, and hypertension. Additionally, we found a decrease in renal blood flow and an increase in renal vascular resistance. Biochemical markers of chronic kidney disease were detected; there was an infiltration of inflammatory cells with an elevated expression of proinflammatory cytokines (tumor necrosis factor-α, interleukin (IL)-6, and IL-1β), the activation of the renin–angiotensin system, and oxidative/nitrosative stress. Notably, in rats exposed to the DH diet for 120 days, the concomitant treatment with isoflavones after 60 days was able to revert metabolic parameters, renal alterations, and oxidative/nitrosative stress. The beneficial effects of isoflavones in the kidney of the obese rats were found to be mediated by expression of peroxisome proliferator-activated receptor gamma (PPAR-γ).
Collapse
Affiliation(s)
- Edson de Andrade Pessoa
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
| | - Márcia Bastos Convento
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
| | - Bianca Castino
- Interdisciplinary Postgraduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo SP 01506-000, Brazil; (B.C.); (A.A.)
| | - Ala Moana Leme
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
| | - Andréia Silva de Oliveira
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
| | - Alef Aragão
- Interdisciplinary Postgraduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo SP 01506-000, Brazil; (B.C.); (A.A.)
| | - Sheila Marques Fernandes
- Experimentation Laboratory in Animal Model, School of Nursing, Universidade de São Paulo, São Paulo SP 05403-000, Brazil; (S.M.F.); (C.D.)
| | - Adriana Carbonel
- Histology and Structural Biology Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo SP 04039-032, Brazil;
| | - Cassiane Dezoti
- Experimentation Laboratory in Animal Model, School of Nursing, Universidade de São Paulo, São Paulo SP 05403-000, Brazil; (S.M.F.); (C.D.)
| | - Maria de Fátima Vattimo
- Department Medical-Surgical Nursing, School of Nursing, Universidade de São Paulo, São Paulo SP 05403-000, Brazil;
| | - Nestor Schor
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
| | - Fernanda Teixeira Borges
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo SP 04023-900, Brazil; (E.d.A.P.); (M.B.C.); (A.M.L.); (A.S.d.O.); (N.S.)
- Interdisciplinary Postgraduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo SP 01506-000, Brazil; (B.C.); (A.A.)
- Correspondence: ; Tel.: +55-11-5576-4242
| |
Collapse
|
36
|
Pei K, Gui T, Li C, Zhang Q, Feng H, Li Y, Wu J, Gai Z. Recent Progress on Lipid Intake and Chronic Kidney Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3680397. [PMID: 32382547 PMCID: PMC7196967 DOI: 10.1155/2020/3680397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022]
Abstract
The incidence of chronic kidney disease (CKD) is associated with major abnormalities in circulating lipoproteins and renal lipid metabolism. This article elaborates on the mechanisms of CKD and lipid uptake abnormalities. The viewpoint we supported is that lipid abnormalities directly cause CKD, resulting in forming a vicious cycle. On the theoretical and experiment fronts, this inference has been verified by elaborately elucidating the role of lipid intake and accumulation as well as their influences on CKD. Taken together, these findings suggest that further understanding of lipid metabolism in CKD may lead to novel therapeutic approaches.
Collapse
Affiliation(s)
- Ke Pei
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ting Gui
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chao Li
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qian Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huichao Feng
- Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhibo Gai
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
37
|
Szczuko M, Kaczkan M, Małgorzewicz S, Rutkowski P, Dębska-Ślizień A, Stachowska E. The C18:3n6/C22:4n6 ratio is a good lipid marker of chronic kidney disease (CKD) progression. Lipids Health Dis 2020; 19:77. [PMID: 32303226 PMCID: PMC7164198 DOI: 10.1186/s12944-020-01258-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
Background Chronic kidney disease (CKD) is a major challenge for public health due to increased risk of cardiovascular diseases (CVD) and premature death. The aim of this study was to determine the clinical picture of FA and the course of the pathophysiological mechanisms of CKD. Methods The study involved 149 patients with CKD and a control group including 43 people. Fatty acid profiles were investigated using gas chromatography. A total of 30 fatty acids and their derivatives were identified and quantified. The omega3, omega6, SFA, MUFA, and PUFA fatty acid contents were calculated. The correlation matrix was obtained for parameters relating to patients with CKD vs. FA, taking patients’ sex into consideration. The index C18:3n6/C22:4n6 was calculated according to the length of the treatment. Statistica 12.0 software (Tulsa, Oklahoma, USA) was used for the statistical analyses. Results The results showed decreased levels of total PUFA and increased concentrations of MUFA, including the activation of the palmitic and oleic acid pathway. An increase in the levels of n-6 9C22: 4n6 family fatty acids in all the patients and a reduction in the n-3 family (EPA, DHA) were observed. C18:3n6 was negatively correlated and C22:4n6 was positively correlated with the duration of the treatment. The index C18:3n6/C22:4n6 was defined as a new marker in the progression of the disease. Moreover, the index C18:3n6/ C22:4n6 was drastically decreased in later period. Nervonic acid was higher in the CKD group. In the group of men with CKD, there was a negative correlation between the excretion of K+, anthropometric measurements, and the levels of EPA and DHA. Conclusions The course of inflammation in CKD occurs through the decrease in PUFA and the synthesis of MUFA. The dominating cascade of changes is the elongation of GLA-C18:3n6 into DGLA-C20:3n6 and AA-C20:4n6. As CKD progresses, along with worsening anthropometrical parameters and increased secretion of potassium, the activity of Ʌ6-desaturase decreases, reducing the synthesis of EPA and DHA. The synthesis of AdA-C22:4n6 increases and the ratio C18:3n6/C22:4n6 drastically decreases after 5 years. This parameter can be used to diagnose disease progression.
Collapse
Affiliation(s)
- Małgorzata Szczuko
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| | - Małgorzata Kaczkan
- Department of Clinical Nutrition and Dietetics, Medical University of Gdańsk, Gdańsk, Poland
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition and Dietetics, Medical University of Gdańsk, Gdańsk, Poland
| | - Przemysław Rutkowski
- Department of General Nursery, Medical University of Gdańsk and Diaverum Hemodialysis Unit, Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
38
|
Opazo-Ríos L, Mas S, Marín-Royo G, Mezzano S, Gómez-Guerrero C, Moreno JA, Egido J. Lipotoxicity and Diabetic Nephropathy: Novel Mechanistic Insights and Therapeutic Opportunities. Int J Mol Sci 2020; 21:E2632. [PMID: 32290082 PMCID: PMC7177360 DOI: 10.3390/ijms21072632] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023] Open
Abstract
Lipotoxicity is characterized by the ectopic accumulation of lipids in organs different from adipose tissue. Lipotoxicity is mainly associated with dysfunctional signaling and insulin resistance response in non-adipose tissue such as myocardium, pancreas, skeletal muscle, liver, and kidney. Serum lipid abnormalities and renal ectopic lipid accumulation have been associated with the development of kidney diseases, in particular diabetic nephropathy. Chronic hyperinsulinemia, often seen in type 2 diabetes, plays a crucial role in blood and liver lipid metabolism abnormalities, thus resulting in increased non-esterified fatty acids (NEFA). Excessive lipid accumulation alters cellular homeostasis and activates lipogenic and glycogenic cell-signaling pathways. Recent evidences indicate that both quantity and quality of lipids are involved in renal damage associated to lipotoxicity by activating inflammation, oxidative stress, mitochondrial dysfunction, and cell-death. The pathological effects of lipotoxicity have been observed in renal cells, thus promoting podocyte injury, tubular damage, mesangial proliferation, endothelial activation, and formation of macrophage-derived foam cells. Therefore, this review examines the recent preclinical and clinical research about the potentially harmful effects of lipids in the kidney, metabolic markers associated with these mechanisms, major signaling pathways affected, the causes of excessive lipid accumulation, and the types of lipids involved, as well as offers a comprehensive update of therapeutic strategies targeting lipotoxicity.
Collapse
Affiliation(s)
- Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (L.O.-R.); (G.M.-R.); (C.G.-G.); (J.E.)
| | - Sebastián Mas
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (L.O.-R.); (G.M.-R.); (C.G.-G.); (J.E.)
| | - Gema Marín-Royo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (L.O.-R.); (G.M.-R.); (C.G.-G.); (J.E.)
| | - Sergio Mezzano
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, 5090000 Valdivia, Chile;
| | - Carmen Gómez-Guerrero
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (L.O.-R.); (G.M.-R.); (C.G.-G.); (J.E.)
| | - Juan Antonio Moreno
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, 14004 Cordoba, Spain
- Hospital Universitario Reina Sofía, 14004 Cordoba, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain; (L.O.-R.); (G.M.-R.); (C.G.-G.); (J.E.)
| |
Collapse
|
39
|
Zhou ZF, Jiang L, Zhao Q, Wang Y, Zhou J, Chen QK, Lv JL. Roles of pattern recognition receptors in diabetic nephropathy. J Zhejiang Univ Sci B 2020; 21:192-203. [PMID: 32133797 DOI: 10.1631/jzus.b1900490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diabetic nephropathy (DN) is currently the most common complication of diabetes. It is considered to be one of the leading causes of end-stage renal disease (ESRD) and affects many diabetic patients. The pathogenesis of DN is extremely complex and has not yet been clarified; however, in recent years, increasing evidence has shown the important role of innate immunity in DN pathogenesis. Pattern recognition receptors (PRRs) are important components of the innate immune system and have a significant impact on the occurrence and development of DN. In this review, we classify PRRs into secretory, endocytic, and signal transduction PRRs according to the relationship between the PRRs and subcellular compartments. PRRs can recognize related pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), thus triggering a series of inflammatory responses, promoting renal fibrosis, and finally causing renal impairment. In this review, we describe the proposed role of each type of PRRs in the development and progression of DN.
Collapse
Affiliation(s)
- Zhi-Feng Zhou
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Lei Jiang
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| | - Qing Zhao
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| | - Yu Wang
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| | - Jing Zhou
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| | - Qin-Kai Chen
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| | - Jin-Lei Lv
- Department of Nephrology, the First Affiliated Hospital of Nanchang University, Institute of Molecular Immunology of Kidney Disease of Nanchang University, Nanchang 330006, China
| |
Collapse
|
40
|
Hou W, Liu B, Xu H. Celastrol: Progresses in structure-modifications, structure-activity relationships, pharmacology and toxicology. Eur J Med Chem 2020; 189:112081. [DOI: 10.1016/j.ejmech.2020.112081] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
|
41
|
McPherson KC, Shields CA, Poudel B, Johnson AC, Taylor L, Stubbs C, Nichols A, Cornelius DC, Garrett MR, Williams JM. Altered renal hemodynamics is associated with glomerular lipid accumulation in obese Dahl salt-sensitive leptin receptor mutant rats. Am J Physiol Renal Physiol 2020; 318:F911-F921. [PMID: 32068459 DOI: 10.1152/ajprenal.00438.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The present study examined whether development of renal injury in the nondiabetic obese Dahl salt-sensitive leptin receptor mutant (SSLepRmutant) strain is associated with elevations in glomerular filtration rate and renal lipid accumulation. Baseline mean arterial pressure at 6 wk of age was similar between Dahl salt-sensitive wild-type (SSWT) and SSLepRmutant rats. However, by 18 wk of age, the SSLepRmutant strain developed hypertension, while the elevation in mean arterial pressure was not as severe in SSWT rats (192 ± 4 and 149 ± 6 mmHg, respectively). At baseline, proteinuria was fourfold higher in SSLepRmutant than SSWT rats and remained elevated throughout the study. The early development of progressive proteinuria was associated with renal hyperfiltration followed by a decline in renal function over the course of study in the SSLepRmutant compared with SSWT rats. Kidneys from the SSLepRmutant strain displayed more glomerulosclerosis and glomerular lipid accumulation than SSWT rats. Glomeruli were isolated from the renal cortex of both strains at 6 and 18 wk of age, and RNA sequencing was performed to identify genes and pathways driving glomerular injury. We observed significant increases in expression of the influx lipid transporters, chemokine (C-X-C motif) ligand 16 (Cxcl16) and scavenger receptor and fatty acid translocase (Cd36), respectively, and a significant decrease in expression of the efflux lipid transporter, ATP-binding cassette subfamily A member 2 (Abca2; cholesterol efflux regulatory protein 2), in SSLepRmutant compared with SSWT rats at 6 and 18 wk of age, which were validated by RT-PCR analysis. These data suggest an association between glomerular hyperfiltration and glomerular lipid accumulation during the early development of proteinuria associated with obesity.
Collapse
Affiliation(s)
- Kasi C McPherson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bibek Poudel
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ashley C Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lateia Taylor
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cassandra Stubbs
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alyssa Nichols
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
42
|
Ferrara D, Montecucco F, Dallegri F, Carbone F. Impact of different ectopic fat depots on cardiovascular and metabolic diseases. J Cell Physiol 2019; 234:21630-21641. [PMID: 31106419 DOI: 10.1002/jcp.28821] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
A growing body of evidence is pointing out the pathophysiological role of fat accumulation in different organs. Ectopic fat depots within heart, liver, skeletal muscle, kidney, and pancreas as well as around blood vessels might be more associated to cardiometabolic risk than classical variables, such as body mass index. Among different mechanisms, lipid metabolism appears to be particularly influenced by ectopic fat depots. Indeed, intracellular accumulation of nonesterified fatty acids, and triglycerides promotes endoplasmic reticulum stress, mitochondrial uncoupling, oxidative stress, and altered membrane composition/function, finally promoting inflammatory response and cell death. The dysfunctional adipose tissue was shown to induce both local and systemic effects, with relevant clinical consequences. Epicardial fat and myocardial steatosis have been associated with the development of atrial fibrillation and ventricular dysfunction. Similarly perivascular adipose tissue appears to trigger atherosclerosis and hypertension. Nonalcoholic fatty liver disease has been recognized both as the hepatic manifestation of metabolic syndrome and as a cardiovascular (CV) risk factor. Importantly, the renal sinus fat emerged as a potential player in kidney dysfunction. Finally, both skeletal muscle and pancreatic fat depots have been indicated as potential endocrine modulators of insulin resistance. Considering the global rise in the prevalence of obesity, the understanding of mechanisms underlying ectopic fat accumulation represents an urgent need, with potential clinical implications for CV risk stratification. Here, we attempt to update the current knowledge of the different ectopic fat depots, focusing on underlying mechanisms and potential clinical implications.
Collapse
Affiliation(s)
- Daniele Ferrara
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Centre of Excellence for Biomedical Research (CEBR), Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Franco Dallegri
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
43
|
Wu Z, Ma Y, Gong X, Zhang Y, Zhao L, Cheng G, Cai S. Rhus chinensis Mill. fruits prevent high-fat/ethanol diet-induced alcoholic fatty liver in rats via AMPK/SREBP-1/FAS signaling pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103498] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
44
|
Yang H, Li H. CD36 identified by weighted gene co-expression network analysis as a hub candidate gene in lupus nephritis. PeerJ 2019; 7:e7722. [PMID: 31592160 PMCID: PMC6777479 DOI: 10.7717/peerj.7722] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022] Open
Abstract
Lupus nephritis (LN) is a severe manifestation of systemic lupus erythematosus (SLE), which often progresses to end-stage renal disease (ESRD) and ultimately leads to death. At present, there are no definitive therapies towards LN, so that illuminating the molecular mechanism behind the disease has become an urgent task for researchers. Bioinformatics has become a widely utilized method for exploring genes related to disease. This study set out to conduct weighted gene co-expression network analysis (WGCNA) and screen the hub gene of LN. We performed WGCNA on the microarray expression profile dataset of GSE104948 from the Gene Expression Omnibus (GEO) database with 18 normal and 21 LN samples of glomerulus. A total of 5,942 genes were divided into 5 co-expression modules, one of which was significantly correlated to LN. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted on the LN-related module, and the module was proved to be associated mainly with the activation of inflammation, immune response, cytokines, and immune cells. Genes in the most significant GO terms were extracted for sub-networks of WGNCA. We evaluated the centrality of genes in the sub-networks by Maximal Clique Centrality (MCC) method and CD36 was ultimately screened out as a hub candidate gene of the pathogenesis of LN. The result was verified by its differentially expressed level between normal and LN in GSE104948 and the other three multi-microarray datasets of GEO. Moreover, we further demonstrated that the expression level of CD36 is related to the WHO Lupus Nephritis Class of LN patients with the help of Nephroseq database. The current study proposed CD36 as a vital candidate gene in LN for the first time and CD36 may perform as a brand-new biomarker or therapeutic target of LN in the future.
Collapse
Affiliation(s)
- Huiying Yang
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Hua Li
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
45
|
FcgRIII Deficiency and FcgRIIb Defeciency Promote Renal Injury in Diabetic Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3514574. [PMID: 31534958 PMCID: PMC6724446 DOI: 10.1155/2019/3514574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/12/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023]
Abstract
The immune system is involved in the development of diabetes complications and IgG Fc gamma receptors (FcgRs) are key immune receptors responsible for the effective control of both humoral and innate immunity. We investigated the effects of members of the FcgR superfamily into both the streptozotocin plus high fat-induced type 2 diabetes and high fat diet (HFD) models. FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice had elevated levels of serum creatinine compared with wildtype (WT) diabetic mice. Renal histology of diabetic FcgRIII knockout and FcgRIIb knockout mice showed mesangial expansion and GBM thickening; the mechanistic study indicated a higher expression of TGF-β1, TNF-α, and p-NFκB-p65 compared with wild type mouse. The HFD mouse with FcgRIII knockout or FcgRIIb knockout had increased biochemical and renal injury factors, but oxLDL deposition was higher than in FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice. In vitro we further examined the mechanism by which the Fc gamma receptor promoted renal injury and transfected glomerular mesangial cells (GMCs) with FcgRI siRNA attenuated the level of TGF-β1, TNF-α expression. In summary, FcgRI knockdown downregulated kidney inflammation and fibrosis and FcgRIIb knockout accelerated inflammation, fibrosis, and the anomalous deposition of oxLDL whereas FcgRIII deficiency failed to protect kidney from diabetic renal injury. These observations suggested that FcgRs might represent a novel target for the therapeutic intervention of diabetic nephropathy.
Collapse
|
46
|
Muller CR, Leite APO, Yokota R, Pereira RO, Americo ALV, Nascimento NRF, Evangelista FS, Farah V, Fonteles MC, Fiorino P. Post-weaning Exposure to High-Fat Diet Induces Kidney Lipid Accumulation and Function Impairment in Adult Rats. Front Nutr 2019; 6:60. [PMID: 31131281 PMCID: PMC6509178 DOI: 10.3389/fnut.2019.00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/15/2019] [Indexed: 12/24/2022] Open
Abstract
Aim: We investigated the kidney morphofunctional consequences of high-fat diet intake since post-weaning in adult rats. Main Methods: Male Wistar rats were divided into two groups: ND (normal diet; n = 10) and HD (high-fat diet; n = 10). The high-fat diet was introduced post-weaned and animals were followed for 8 weeks. Key Findings: HD group did not change body weight gain even though food consumption has decreased with no changes in caloric consumption. The HD group showed glucose intolerance and insulin resistance. The glomerular filtration rate (GFR) was decreased in vivo (ND: 2.8 ± 1.01; HD: 1.1 ± 0.14 ml/min) and in the isolated perfusion method (34% of decrease). Renal histological analysis showed a retraction in glomeruli and an increase in kidney lipid deposition (ND: 1.5 ± 0.17 HD: 5.9 ± 0.06%). Furthermore, the high-fat diet consumption increased the pro-inflammatory cytokines IL-6 (ND: 1,276 ± 203; HD: 1,982 ± 47 pg/mL/mg) and IL-1b (ND: 97 ± 12 HD: 133 ± 5 pg/mL/mg) without changing anti-inflammatory cytokine IL-10. Significance: Our study provides evidence that high-fat diet consumption leads to renal lipid accumulation, increases inflammatory cytokines, induces glomeruli retraction, and renal dysfunction. These damages observed in the kidney could be associated with an increased risk to advanced CKD in adulthood suggesting that reduction of high-fat ingestion during an early period of life can prevent metabolic disturbances and renal lipotoxicity.
Collapse
Affiliation(s)
- Cynthia R Muller
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Experimental Pathophysiology Department, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Ana Paula O Leite
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Department of Medicine, Renal Division, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo Yokota
- Department of Medicine, Renal Division, Federal University of São Paulo, São Paulo, Brazil
| | - Renata O Pereira
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Department of Medicine, Translational Medicine Division, Federal University of São Paulo, São Paulo, Brazil
| | - Anna Laura V Americo
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Experimental Pathophysiology Department, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | - Vera Farah
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Department of Medicine, Translational Medicine Division, Federal University of São Paulo, São Paulo, Brazil
| | - Manasses C Fonteles
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil.,Superior Institute of Biomedical Sciences, Ceara State University, Fortaleza, Brazil
| | - Patricia Fiorino
- Renal, Cardiovascular and Metabolic Physiopharmacology Laboratory, Health and Biological Science Center, Mackenzie Presbyterian University, São Paulo, Brazil
| |
Collapse
|
47
|
Definition of an oxidative stress status by combined assessment of Malondialdehyde and Oxidized-LDL: A study in patients with type2 diabetes and control. Meta Gene 2019. [DOI: 10.1016/j.mgene.2018.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
48
|
Ng SW, Chan Y, Chellappan DK, Madheswaran T, Zeeshan F, Chan YL, Collet T, Gupta G, Oliver BG, Wark P, Hansbro N, Hsu A, Hansbro PM, Dua K, Panneerselvam J. Molecular modulators of celastrol as the keystones for its diverse pharmacological activities. Biomed Pharmacother 2019; 109:1785-1792. [DOI: 10.1016/j.biopha.2018.11.051] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/10/2018] [Accepted: 11/10/2018] [Indexed: 12/30/2022] Open
|
49
|
Furukawa S, Suzuki H, Fujihara K, Kobayashi K, Iwasaki H, Sugano Y, Yatoh S, Sekiya M, Yahagi N, Shimano H. Malondialdehyde-modified LDL-related variables are associated with diabetic kidney disease in type 2 diabetes. Diabetes Res Clin Pract 2018; 141:237-243. [PMID: 29775676 DOI: 10.1016/j.diabres.2018.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/03/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Oxidized low-density lipoprotein (oxLDL) causes the development of atherosclerosis and kidney injury. Although circulating oxLDL levels were reportedly increased in type 2 diabetic patients with macroalbuminuria, it remains unclear whether albuminuria or the reduced glomerular filtration rate (GFR) is independently associated with the circulating oxLDL level. This study aimed to elucidate the association between the stage of diabetic nephropathy and serum malondialdehyde-modified LDL (MDA-LDL) and the ratio of MDA-LDL to LDL-cholesterol (MDA-LDL/LDL). METHODS AND RESULTS This retroactive cross-sectional study used data from 402 patients with type 2 diabetes. Patients undergoing hemodialysis were excluded. Serum MDA-LDL levels were significantly increased with increases in severity of albuminuria (103 ± 44 U/L, 109 ± 54 U/L, and 135 ± 72 U/L for normoalbuminuria, microalbuminuria, and macroalbuminuria, respectively; P for trend = 0.020) but not according to the estimated GFR (eGFR). An increased MDA-LDL/LDL ratio was significantly associated with both increased albuminuria (35 ± 13, 37 ± 14, and 40 ± 15 for normoalbuminuria, microalbuminuria, and macroalbuminuria, respectively; P for trend = 0.003) and reduced eGFR (34 ± 13, 36 ± 13, 38 ± 12, and 51 ± 28 for grade 1, 2, 3 and 4, respectively; P for trend = 0.002). Multiple linear regression analysis showed that neither the albumin excretion rate nor eGFR but ln-transformed triglycerides and LDL-C levels were independent determinants of both serum MDA-LDL levels and MDA-LDL/LDL ratios. CONCLUSION Serum MDA-LDL levels and MDA-LDL/LDL ratios were increased in those with dyslipidemia associated with diabetic kidney disease.
Collapse
Affiliation(s)
- Shoko Furukawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroaki Suzuki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Kazuto Kobayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hitoshi Iwasaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoko Sugano
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shigeru Yatoh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Motohiro Sekiya
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoya Yahagi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hitoshi Shimano
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
50
|
Hu ZB, Ma KL, Zhang Y, Wang GH, Liu L, Lu J, Chen PP, Lu CC, Liu BC. Inflammation-activated CXCL16 pathway contributes to tubulointerstitial injury in mouse diabetic nephropathy. Acta Pharmacol Sin 2018; 39:1022-1033. [PMID: 29620052 DOI: 10.1038/aps.2017.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/19/2017] [Indexed: 02/06/2023]
Abstract
Inflammation and lipid disorders play crucial roles in synergistically accelerating the progression of diabetic nephropathy (DN). In this study we investigated how inflammation and lipid disorders caused tubulointerstitial injury in DN in vivo and in vitro. Diabetic db/db mice were injected with 10% casein (0.5 mL, sc) every other day for 8 weeks to cause chronic inflammation. Compared with db/db mice, casein-injected db/db mice showed exacerbated tubulointerstitial injury, evidenced by increased secretion of extracellular matrix (ECM) and cholesterol accumulation in tubulointerstitium, which was accompanied by activation of the CXC chemokine ligand 16 (CXCL16) pathway. In the in vitro study, we treated HK-2 cells with IL-1β (5 ng/mL) and high glucose (30 mmol/L). IL-1β treatment increased cholesterol accumulation in HK-2 cells, leading to greatly increased ROS production, ECM protein expression levels, which was accompanied by the upregulated expression levels of proteins in the CXCL16 pathway. In contrast, after CXCL16 in HK-2 cells was knocked down by siRNA, the IL-1β-deteriorated changes were attenuated. In conclusion, inflammation accelerates renal tubulointerstitial lesions in mouse DN via increasing the activity of CXCL16 pathway.
Collapse
|