1
|
Xing Y, Huang D, Lin P, Zhou Y, Chen D, Ye C, Wu M. Salvianolic acid C promotes renal gluconeogenesis in fibrotic kidneys through PGC1α. Biochem Biophys Res Commun 2025; 744:151174. [PMID: 39700761 DOI: 10.1016/j.bbrc.2024.151174] [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: 12/04/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Impaired renal gluconeogenesis is recently identified as a hallmark of chronic kidney disease. However, the therapeutic approach to promote renal gluconeogenesis in CKD is still lacking. We aimed to study whether Salvianolic acid C (SAC), a nature compound extracted from the traditional Chinese medicine Danshen, inhibits renal fibrosis through promotion of gluconeogenesis. TGF-β stimulated HK2 human renal epithelial cells and mice with unilateral ureteral obstruction (UUO) were used as in vitro and in vivo models to study renal fibrosis. Fibrotic and gluconeogenic changes were determined by Western blotting analysis, quantitative PCR and Masson staining. Glucose and lactate concentrations were measured in cell culture and renal tissues. We found that SAC treatment inhibits the deposition of extracellular matrix proteins and the expression of fibrotic markers such as fibronectin, N-cadherin, Vimentin, aSMA, pSmad3, and Snail in UUO kidneys or renal cells. Inhibition of these fibrotic markers by SAC treatment was associated with enhanced expression of three gluconeogenic enzymes such as PCK1, G6PC and FBP1 in renal tissues or cells. SAC increase the concentration of glucose in the supernatant of renal cells. Lactate concentration was reduced by SAC in renal tissues or cells. Pyruvate and glucose tolerance tests showed that SAC improve the impaired glucose metabolism systemically in UUO mice. Peroxisome proliferator activated receptor gamma coactivator 1 alpha (PGC1ɑ) was downregulated in mouse kidneys after UUO operation, which was increased by SAC treatment. Moreover, PGC1α inhibitor SR-18292 reversed the anti-fibrotic effect and pro-gluconeogenic effect caused by SAC in renal cells. In conclusion, SAC inhibits renal fibrosis through promotion of PGC1α-mediated renal gluconeogenesis.
Collapse
Affiliation(s)
- Yufeng Xing
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Di Huang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Pinglan Lin
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Yijing Zhou
- Department of Nephrology, JiaXing Hospital of Traditional Chinese Medicine, JiaXing, China
| | - Dongping Chen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China
| | - Chaoyang Ye
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China.
| | - Ming Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, China; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, China; Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, China.
| |
Collapse
|
2
|
Miguel V, Shaw IW, Kramann R. Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nat Rev Nephrol 2025; 21:39-56. [PMID: 39289568 DOI: 10.1038/s41581-024-00889-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
Chronic kidney disease (CKD), defined as persistent (>3 months) kidney functional loss, has a growing prevalence (>10% worldwide population) and limited treatment options. Fibrosis driven by the aberrant accumulation of extracellular matrix is the final common pathway of nearly all types of chronic repetitive injury in the kidney and is considered a hallmark of CKD. Myofibroblasts are key extracellular matrix-producing cells that are activated by crosstalk between damaged tubules and immune cells. Emerging evidence indicates that metabolic alterations are crucial contributors to the pathogenesis of kidney fibrosis by affecting cellular bioenergetics and metabolite signalling. Immune cell functions are intricately connected to their metabolic characteristics, and kidney cells seem to undergo cell-type-specific metabolic shifts in response to damage, all of which can determine injury and repair responses in CKD. A detailed understanding of the heterogeneity in metabolic reprogramming of different kidney cellular subsets is essential to elucidating communication processes between cell types and to enabling the development of metabolism-based innovative therapeutic strategies against CKD.
Collapse
Affiliation(s)
- Verónica Miguel
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Isaac W Shaw
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Rafael Kramann
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany.
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Wang C, Zhou J, Jia P, Yang Y, Song R, Zheng X, Zhang H, Li Y. Joint proteomic and metabolomic analysis reveals renal metabolic remodeling of chronic heart failure mice. J Pharm Biomed Anal 2024; 255:116641. [PMID: 39731929 DOI: 10.1016/j.jpba.2024.116641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
Pharmacologic intervention in chronic heart failure (HF) with renal insufficiency is one of the clinical challenges due to the fact that the mechanisms of cardio-renal interactions in chronic heart failure (CHF) progressing have not been fully revealed. In this paper, C57BL/6 mice were applied thoracic aortic narrowing surgery to establish pressure overload CHF model. Cardiac function, serum markers, renal pathologic changes and kidney metabolism were analyzed at 4th, 8th, 12th, and 16th week after surgery respectively to evaluate the heart-Kidney pathologic overlap. Kidney proteomic analysis was performed at 16th week after operation. As a result, renal hypofiltration and exacerbation of pathological damage was observed accompanying cardiac function deterioration after 12th week. 66 differentially expressed proteins and 13 differential metabolites were found to be involved in the cardio-renal pathological overlap. Joint proteomic and metabolomic analysis revealed that signal pathways like Phosphatidylinositol signaling system, Glucagon signaling pathway, the Glyoxylate and dicarboxylate metabolism; DEPs of Pten, Mtmr4, PLC and CPT1, differential metabolites like aspartic acid and isocitrate deserve further investigation.
Collapse
Affiliation(s)
- Chunliu Wang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Jie Zhou
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Ruixue Song
- Xi'an Research Institute of Chinese Lacquer, Xi'an, Shaanxi, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| | - Ye Li
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Liu Z, Dai B, Bao J, Pan Y. T cell metabolism in kidney immune homeostasis. Front Immunol 2024; 15:1498808. [PMID: 39737193 PMCID: PMC11684269 DOI: 10.3389/fimmu.2024.1498808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Kidney immune homeostasis is intricately linked to T cells. Inappropriate differentiation, activation, and effector functions of T cells lead to a spectrum of kidney disease. While executing immune functions, T cells undergo a series of metabolic rewiring to meet the rapid energy demand. The key enzymes and metabolites involved in T cell metabolism metabolically and epigenetically modulate T cells' differentiation, activation, and effector functions, thereby being capable of modulating kidney immune homeostasis. In this review, we first summarize the latest advancements in T cell immunometabolism. Second, we outline the alterations in the renal microenvironment under certain kidney disease conditions. Ultimately, we highlight the metabolic modulation of T cells within kidney immune homeostasis, which may shed light on new strategies for treating kidney disease.
Collapse
Affiliation(s)
- Zikang Liu
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Binbin Dai
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jiwen Bao
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yangbin Pan
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
5
|
Çevik ZR, Erdoğan A, Mumcu A, Doğan B. Investigation of metabolite profiles of kidney tissues in rats treated with favipiravir drug: An NMR-based metabolomics study. J Pharm Biomed Anal 2024; 255:116640. [PMID: 39705846 DOI: 10.1016/j.jpba.2024.116640] [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: 09/06/2024] [Revised: 11/29/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
In response to the urgent need for effective treatments during the rapid spread and high mortality rate of COVID-19, existing drugs were repurposed for potential antiviral effects, including favipiravir, originally designed as an RNA-dependent RNA polymerase inhibitor for influenza. Despite limited antiviral effectiveness against COVID-19, favipiravir has been reported to cause several adverse drug events (ADEs) in the body. Recent studies have shown that favipiravir can damage various tissues in rats. However, a detailed analysis of its effects on the metabolomics profile of tissues using high-resolution spectroscopic technologies has not yet been conducted. In this study, it was aimed to analyze the metabolomic changes in rat kidney tissues induced by favipiravir, using high-resolution nuclear magnetic resonance (NMR) spectroscopy. Sixty male Wistar Albino rats were randomly divided into three groups: control, low-dose favipiravir (200 mg/kg), and high-dose favipiravir (300 mg/kg), with 20 rats per group. Each group received its respective treatment via oral gavage. After the treatment period, kidney tissue samples were collected and subjected to 1H NMR analysis. Bioinformatics analysis of the obtained 1H NMR spectra suggests that favipiravir induces dose-dependent metabolic changes in kidney tissue, with higher doses causing more profound disruptions in several pathways.
Collapse
Affiliation(s)
| | - Ali Erdoğan
- Department of Biomedical Engineering, Inonu University, Malatya, Turkey
| | - Akın Mumcu
- Laboratory of NMR, Scientific and Technological Research Center, Inonu University, Malatya, Turkey
| | - Berat Doğan
- Department of Biomedical Engineering, Inonu University, Malatya, Turkey.
| |
Collapse
|
6
|
Xiang T, Wang X, Huang S, Zhou K, Fei S, Zhou B, Yue K, Li Q, Xue S, Dai Y, Zhang J, Ni H, Sun C, Huang X. Inhibition of PKM2 by shikonin impedes TGF-β1 expression by repressing histone lactylation to alleviate renal fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 136:156324. [PMID: 39700636 DOI: 10.1016/j.phymed.2024.156324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Macrophage-myofibroblast transition (MMT) plays a significant role in the progression of renal fibrosis in chronic kidney disease (CKD), making inhibition of MMT a promising therapeutic strategy. Pyruvate kinase M2 (PKM2) and its metabolite lactate are implicated in the pathogenesis of renal fibrosis; however, the mechanisms through which they contribute to this process remain poorly understood. PURPOSE To investigate the effects of PKM2 inhibition by shikonin on renal fibrosis and the underly mechanisms. METHODS Mice were subjected to unilateral ureteral obstruction (UUO) to establish a CKD model. Renal fibrosis was assessed using histochemistry and western blotting. The MMT and histone lactylation levels were evaluated by immunofluorescence and western blotting. The interaction between the Tgfb1 promoter and lactylated histone H3 (K18) was examined using chromatin Immunoprecipitation (ChIP). RESULTS PKM2 expression was significantly elevated in the renal tubular cells of UUO mouse kidneys, resulting in increased pyruvate and lactate production. Similarly, lactate levels were elevated in TGF-β1-treated TCMK-1 cells and in the serum of CKD patients. In UUO mice, treatment with shikonin, a potent PKM2 inhibitor, effectively reduced lactate production, alleviated renal fibrosis, decreased TGF-β1 expression, and suppressed the MMT process. Mechanistic studies revealed that lactate treatment stimulates Tgfb1 expression in TCMK-1 cells. Consequently, TGF-β1 in conditioned media from lactate-treated TCMK-1 cells promoted M2 macrophage polarization and upregulated fibrotic gene expression in RAW264.7 cells. Pharmacological intervention demonstrated that TGF-β1 activates the Smad3 pathway to drive the MMT process. In TCMK-1 cells, both lactate treatment and PKM2 overexpression induced Tgfb1 expression by promoting histone H3K18 lactylation. CONCLUSIONS Our findings indicate that PKM2-induced excessive lactate production renal tubular cells contributes to renal fibrosis. Lactate promotes histone lactylation, leading to TGF-β1 expression in these cells, which subsequently activates the Smad3 pathway in macrophages, driving the MMT and fibrosis in the kidney. Therefore, targeting PKM2, as with shikonin treatment, may represent an effective therapeutic strategy for managing renal fibrosis in CKD.
Collapse
Affiliation(s)
- Tianya Xiang
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Xijian Wang
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Shujiao Huang
- Xinglin College, Nantong University, Nantong, 226001, China
| | - Kexin Zhou
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Shengnan Fei
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Bing Zhou
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Kun Yue
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Qingxin Li
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Shengnan Xue
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Yongyi Dai
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Jing Zhang
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Haoran Ni
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory of Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, China
| | - Xinzhong Huang
- Department of Nephrology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
7
|
Lin Z, Huo H, Huang M, Tao J, Yang Y, Guo J. Fufang Zhenzhu Tiaozhi (FTZ) capsule ameliorates diabetic kidney disease in mice via inhibiting the SGLT2/glycolysis pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118698. [PMID: 39151712 DOI: 10.1016/j.jep.2024.118698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang Zhenzhu Tiaozhi (FTZ) capsule is a hospital preparation of a patented traditional Chinese medicine compound. FTZ has been clinically used for nearly 13 years in the treatment of diabetes and glycolipid metabolic diseases. With the significant benefits of SGLT2 inhibitor in patients with diabetic kidney disease (DKD), it provides a research avenue to explore the mechanism of FTZ in treating this disease based on glycolysis pathway. AIM OF THE STUDY To explore the pharmacological characteristics of FTZ in DKD mice and its impact on the glycolysis pathway. MATERIALS AND METHODS We induced a DKD model in C57BL/6 mice by injection of streptozotocin (STZ) combined with long-term high-fat diet. We administered three doses of FTZ for 12 weeks of treatment. Kidney function, blood lipid levels, glucose tolerance, and key glycolytic enzymes were evaluated. Renal pathological changes were observed using HE, MASSON, and PAS staining. The potential targets of the active ingredients of FTZ in the glycolysis pathway were predicted using network pharmacology and molecular docking. Validation was performed using immunohistochemistry and Western blotting. RESULTS FTZ effectively reduces blood glucose, total cholesterol, triglyceride, low density lipoprotein cholesterol, 24 h proteinuria, serum creatinine, blood urea nitrogen, and increases urinary glucose levels. Glucose tolerance and renal pathological changes were significantly improved by FTZ treatment. Pinusolidic acid, a component of FTZ, shows good binding affinity with three active pockets of SGLT2. WB and immunohistochemistry revealed that FTZ significantly inhibits the expression of SGLT2 and its glycolytic related proteins (GLUT2/PKM2/HK2). Hexokinase, pyruvate kinase, and lactate dehydrogenase in the kidney were also significantly inhibited by FTZ in a dose-dependent manner. CONCLUSION FTZ may alleviate the progression of DKD by inhibiting the activation of the SGLT2/glycolytic pathway. Our study provides new insights into the clinical application of FTZ in DKD.
Collapse
Affiliation(s)
- Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China
| | - Minyi Huang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China
| | - Jie Tao
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China.
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega Centre, Guangzhou, PR China.
| |
Collapse
|
8
|
Bai Y, Wen H, Lin J, Liu X, Yu H, Wu M, Wang L, Chen D. Tanshinone I improves renal fibrosis by promoting gluconeogenesis through upregulation of peroxisome proliferator-activated receptor-γ coactivator 1α. Ren Fail 2024; 46:2433710. [PMID: 39648664 PMCID: PMC11632924 DOI: 10.1080/0886022x.2024.2433710] [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: 09/04/2024] [Revised: 10/12/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Renal fibrosis, a hallmark of chronic kidney disease, is closely associated with dysregulated gluconeogenesis. Tanshinone I (Tan I), a bioactive compound derived from the traditional Chinese medicine Danshen, exhibits antifibrotic and anti-inflammatory properties. However, its effects on gluconeogenesis and the mechanisms through which it alleviates renal fibrosis remain unclear. This study aimed to investigate whether Tan I promotes gluconeogenesis and mitigates renal fibrosis. METHODS Both in vivo and in vitro experiments were conducted. A unilateral ureteral obstruction (UUO) mouse model was used. Masson's trichrome, HE, and immunofluorescence staining, along with Western blotting, were employed. Lactate concentrations and a pyruvate tolerance test were conducted to assess glucose metabolism. In vitro, HK2 cells and primary renal tubular cells were treated with transforming growth factor-β (TGFβ) to induce fibrosis, and the effects of Tan I on glucose and lactate levels were examined. RESULTS In the UUO model, Tan I reduced fibrosis, decreased lactate accumulation, and modulated fibrosis markers while upregulating gluconeogenesis markers. Tanshinone I restored impaired renal gluconeogenesis, as evidenced by increased pyruvate levels. In vitro, Tan I inhibited fibrosis, reduced lactate levels, and increased glucose levels in cell supernatants. It also restored gluconeogenesis protein expression and decreased fibrotic protein levels. Peroxisome proliferator-activated receptor-γ coactivator (PGC1α) expression was downregulated in UUO and TGFβ-stimulated models, and Tan I reversed this downregulation. Inhibition of PGC1α in TGFβ-stimulated cells counteracted the antifibrotic and gluconeogenesis-promoting effects of Tan I. CONCLUSIONS Tanshinone I ameliorated renal fibrosis by enhancing gluconeogenesis through upregulation of PGC1α.
Collapse
Affiliation(s)
- Yanfang Bai
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Hui Wen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Junyan Lin
- The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinying Liu
- Department of Rheumatology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Hua Yu
- Shanghai Zhabei District Central Hospital, Shanghai, China
| | - Ming Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Dongping Chen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| |
Collapse
|
9
|
Selvarajah V, Robertson D, Hansen L, Jermutus L, Smith K, Coggi A, Sánchez J, Chang YT, Yu H, Parkinson J, Khan A, Chung HS, Hess S, Dumas R, Duck T, Jolly S, Elliott TG, Baker J, Lecube A, Derwahl KM, Scott R, Morales C, Peters C, Goldenberg R, Parker VER, Heerspink HJL. A randomized phase 2b trial examined the effects of the glucagon-like peptide-1 and glucagon receptor agonist cotadutide on kidney outcomes in patients with diabetic kidney disease. Kidney Int 2024; 106:1170-1180. [PMID: 39218393 DOI: 10.1016/j.kint.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Cotadutide is a glucagon-like peptide-1 (GLP-1) and glucagon receptor agonist that may improve kidney function in patients with type 2 diabetes (T2D) and chronic kidney disease (CKD). In this phase 2b study, patients with T2D and CKD (estimated glomerular filtration rate [eGFR] of 20 or more and under 90 mL/min per 1.73 m2 and urinary albumin-to-creatinine ratio [UACR] over 50 mg/g) were randomized 1:1:1:1:1 to 26 weeks' treatment with standard of care plus subcutaneous cotadutide uptitrated to 100, 300, or 600 μg, or placebo daily (double-blind), or the GLP-1 agonist semaglutide 1 mg once weekly (open-label).The co-primary endpoints were absolute and percentage change versus placebo in UACR from baseline to the end of week 14. Among 248 randomized patients, mean age 67.1 years, 19% were female, mean eGFR was 55.3 mL/min per 1.73 m2, geometric mean was UACR 205.5 mg/g (coefficient of variation 270.0), and 46.8% were receiving concomitant sodium-glucose co-transporter 2 inhibitors. Cotadutide dose-dependently reduced UACR from baseline to the end of week 14, reaching significance at 300 μg (-43.9% [95% confidence interval -54.7 to -30.6]) and 600 μg (-49.9% [-59.3 to -38.4]) versus placebo; with effects sustained at week 26. Serious adverse events were balanced across arms. Safety and tolerability of cotadutide 600 μg were comparable to semaglutide. Thus, our study shows that in patients with T2D and CKD, cotadutide significantly reduced UACR on top of standard of care with an acceptable tolerability profile, suggesting kidney protective benefits that need confirmation in a larger study.
Collapse
Affiliation(s)
- Viknesh Selvarajah
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Darren Robertson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Lars Hansen
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Lutz Jermutus
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kirsten Smith
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Angela Coggi
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - José Sánchez
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yi-Ting Chang
- Oncology Biometrics, late Oncology R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Hongtao Yu
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Joanna Parkinson
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anis Khan
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - H Sophia Chung
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Sonja Hess
- Dynamic Omics, Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Richard Dumas
- CISSS Laval, Medecine Department, Endocrinology Division, Laval, Quebec, Canada; Centre de recherches cliniques de Laval, Laval, Quebec, Canada
| | - Tabbatha Duck
- Division of Nephrology, Clinical Research Solutions, Waterloo, Ontario, Canada
| | - Simran Jolly
- Department of Arts and Science, McMaster University, Hamilton, Ontario, Canada
| | | | - John Baker
- Aoteoroa Clinical Trials, Middlemore Hospital, Auckland, New Zealand
| | - Albert Lecube
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital and Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, University of Lleida, Lleida, Catalonia, Spain
| | - Karl-Michael Derwahl
- Institute for Clinical Research and Development, Practise of Endocrinology, Berlin, Germany
| | | | | | - Carl Peters
- Diabetes Services, Te Whatu Ora Waitemata, Auckland, New Zealand
| | | | - Victoria E R Parker
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; The George Institute for Global Health, Sydney, Australia.
| |
Collapse
|
10
|
Vallon V. State-of-the-Art-Review: Mechanisms of Action of SGLT2 Inhibitors and Clinical Implications. Am J Hypertens 2024; 37:841-852. [PMID: 39017631 PMCID: PMC11471837 DOI: 10.1093/ajh/hpae092] [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: 07/11/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Inhibitors of the Na+-coupled glucose transporter SGLT2 (SGLT2i) primarily shift the reabsorption of large amounts of glucose from the kidney's early proximal tubule to downstream tubular segments expressing SGLT1, and the non-reabsorbed glucose is spilled into the urine together with some osmotic diuresis. How can this protect the kidneys and heart from failing as observed in individuals with and without type 2 diabetes? GOAL Mediation analyses identified clinical phenotypes of SGLT2i associated with improved kidney and heart outcome, including a reduction of plasma volume or increase in hematocrit, and lowering of serum urate levels and albuminuria. This review outlines how primary effects of SGLT2i on the early proximal tubule can explain these phenotypes. RESULTS The physiology of tubule-glomerular communication provides the basis for acute lowering of GFR and glomerular capillary pressure, which contributes to lowering of albuminuria but also to long term preservation of GFR, at least in part by reducing kidney cortex oxygen demand. Functional co-regulation of SGLT2 with other sodium and metabolite transporters in the early proximal tubule explains why SGLT2i initially excrete more sodium than expected and are uricosuric, thereby reducing plasma volume and serum urate. Inhibition of SGLT2 reduces early proximal tubule gluco-toxicity and by shifting transport downstream may simulate "systemic hypoxia", and the resulting increase in erythropoiesis, together with the osmotic diuresis, enhances hematocrit and improves blood oxygen delivery. Cardio-renal protection by SGLT2i is also provided by a fasting-like and insulin-sparing metabolic phenotype and, potentially, by off-target effects on the heart and microbiotic formation of uremic toxins.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
11
|
Pan S, Yuan T, Xia Y, Yu W, Li H, Rao T, Ye Z, Li L, Zhou X, Cheng F. SMYD2 Promotes Calcium Oxalate-Induced Glycolysis in Renal Tubular Epithelial Cells via PTEN Methylation. Biomedicines 2024; 12:2279. [PMID: 39457592 PMCID: PMC11504487 DOI: 10.3390/biomedicines12102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Damage to renal tubular cells (RTCs) represents a critical pathological manifestation in calcium oxalate (CaOx) stone disease, but the underlying mechanism remains elusive. Energy metabolism reprogramming is a vital influencer of RTC survival, and SMYD2 is a histone methylation transferase that has been extensively implicated in various metabolic disorders. Hence, this research aimed to identify whether SMYD2 induces the reprogramming of energy metabolism in RTCs exposed to CaOx nephrolithiasis. Methods: Kidney samples were obtained from patients who underwent laparoscopic nephrectomy for non-functioning kidneys caused by nephrolithiasis. The glyoxylate-induced CaOx stone mice model was established and treated with AZ505. The SMYD2-knockout HK-2 cell line was constructed. Histological changes were evaluated by HE, VK, Tunel, Masson stainings. The molecular mechanism was explored through co-immunoprecipitation and western blotting. Results: The results found that SMYD2 upregulation led to energy reprogramming to glycolysis in human kidney tissue samples and in mice with CaOx nephrolithiasis. We also identified the substantial involvement of glycolysis in the induction of apoptosis, inflammation, and epithelial-mesenchymal transition (EMT) in HK-2 cells caused by calcium oxalate monohydrate (COM). In vivo and in vitro results demonstrated that SMYD2 inhibition reduces glycolysis, kidney injury, and fibrosis. Mechanistically, SMYD2 was found to promote metabolic reprogramming of RTCs toward glycolysis by activating the AKT/mTOR pathway via methylated PTEN, which mediates CaOx-induced renal injury and fibrosis. Conclusions: Our findings reveal an epigenetic regulatory role of SMYD2 in metabolic reprogramming in CaOx nephrolithiasis and associated kidney injury, suggesting that targeting SMYD2 and glycolysis may represent a potential therapeutic strategy for CaOx-induced kidney injury and fibrosis.
Collapse
|
12
|
Lumpuy-Castillo J, Amador-Martínez I, Díaz-Rojas M, Lorenzo O, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. Role of mitochondria in reno-cardiac diseases: A study of bioenergetics, biogenesis, and GSH signaling in disease transition. Redox Biol 2024; 76:103340. [PMID: 39250857 PMCID: PMC11407069 DOI: 10.1016/j.redox.2024.103340] [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: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are global health burdens with rising prevalence. Their bidirectional relationship with cardiovascular dysfunction, manifesting as cardio-renal syndromes (CRS) types 3 and 4, underscores the interconnectedness and interdependence of these vital organ systems. Both the kidney and the heart are critically reliant on mitochondrial function. This organelle is currently recognized as a hub in signaling pathways, with emphasis on the redox regulation mediated by glutathione (GSH). Mitochondrial dysfunction, including impaired bioenergetics, redox, and biogenesis pathways, are central to the progression of AKI to CKD and the development of CRS type 3 and 4. This review delves into the metabolic reprogramming and mitochondrial redox signaling and biogenesis alterations in AKI, CKD, and CRS. We examine the pathophysiological mechanisms involving GSH redox signaling and the AMP-activated protein kinase (AMPK)-sirtuin (SIRT)1/3-peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) axis in these conditions. Additionally, we explore the therapeutic potential of GSH synthesis inducers in mitigating these mitochondrial dysfunctions, as well as their effects on inflammation and the progression of CKD and CRS types 3 and 4.
Collapse
Affiliation(s)
- Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - Isabel Amador-Martínez
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico; Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Miriam Díaz-Rojas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 43210, Columbus, Ohio, USA.
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| |
Collapse
|
13
|
Vallon V. How can inhibition of glucose and sodium transport in the early proximal tubule protect the cardiorenal system? Nephrol Dial Transplant 2024; 39:1565-1573. [PMID: 38439675 PMCID: PMC11427065 DOI: 10.1093/ndt/gfae060] [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: 02/04/2024] [Indexed: 03/06/2024] Open
Abstract
What mechanisms can link the inhibition of sodium-glucose cotransporter 2 (SGLT2) in the early proximal tubule to kidney and heart protection in patients with and without type 2 diabetes? Due to physical and functional coupling of SGLT2 to other sodium and metabolite transporters in the early proximal tubule (including NHE3, URAT1), inhibitors of SGLT2 (SGLT2i) reduce reabsorption not only of glucose, inducing osmotic diuresis, but of other metabolites plus of a larger amount of sodium than expected based on SGLT2 inhibition alone, thereby reducing volume retention, hypertension and hyperuricemia. Metabolic adaptations to SGLT2i include a fasting-like response, with enhanced lipolysis and formation of ketone bodies that serve as additional fuel for kidneys and heart. Making use of the physiology of tubulo-glomerular communication, SGLT2i functionally lower glomerular capillary pressure and filtration rate, thereby reducing physical stress on the glomerular filtration barrier, tubular exposure to albumin and nephrotoxic compounds, and the oxygen demand for reabsorbing the filtered load. Together with reduced gluco-toxicity in the early proximal tubule and better distribution of transport work along the nephron, SGLT2i can preserve tubular integrity and transport function and, thereby, glomerular filtration rate in the long-term. By shifting transport downstream, SGLT2i may simulate systemic hypoxia at the oxygen sensors in the deep cortex/outer medulla, which stimulates erythropoiesis and, together with osmotic diuresis, enhances hematocrit and thereby improves oxygen delivery to all organs. The described SGLT2-dependent effects may be complemented by off-target effects of SGLT2i on the heart itself and on the microbiome formation of cardiovascular-effective uremic toxins.
Collapse
Affiliation(s)
- Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
14
|
Tian K, Ang LC, Choudhary P, Choo JCJ, Bee YM, Goh SY, Teh MM. High incidence of low interstitial fluid glucose among type 2 diabetes patients with chronic kidney disease (CKD) despite adhering to appropriate glycated haemoglobin targets-has time come for robust integration of interstitial fluid glucose targets into glycaemic guidelines? Diabet Med 2024:e15438. [PMID: 39301988 DOI: 10.1111/dme.15438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
AIM We aim to compare the burden of Level 1 (<4 mmol/L) and Level 2 (<3 mmol/L) hypoglycaemia between type 2 diabetes (T2D) patients with and without chronic kidney disease (CKD). METHODS T2D subjects with and without CKD (eGFR<60 mL/min/1.73 m2) were recruited from a tertiary-care hospital. Subjects wore the Freestyle Libre-Pro sensor for 2 weeks. The number of hypoglycaemic events and intra-day difference in Level 1 and 2 hypoglycaemias were compared between the cohorts. RESULTS We recruited 134 subjects: 74 with CKD (44 M:30F) and 60 without CKD (36 M:24F), with no difference in HbA1c between the two cohorts (66 ± 20 vs 64 ± 16 mmol/mol, p = 0.529). The CKD cohort had increased level 1 (OR 1.73, p = 0.011), level 2 hypoglycaemias (OR 2.16, p = 0.002), and glycaemic variability than the non-CKD cohort (35.3 ± 9.5 vs 32.3 ± 6.8%). The CKD cohort had more level 2 hypoglycaemia events nocturnally compared to day at 1.9 ± 3.1 vs. 1.4 ± 2.5 events/person within the two week sensor wearing period (p = 0.022), whereas there was no significant intra-day difference in the number of such events within the non-CKD cohort. CONCLUSIONS The CKD cohort has a greater burden of hypoglycaemia despite being treated to similar HbA1c targets. The greater number of nocturnal events warrants safety concern. Interstitial fluid glucose targets should be incorporated into the glycaemic guidelines for T2D patients with CKD.
Collapse
Affiliation(s)
- Kristy Tian
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Li Chang Ang
- Medicine Academic Clinical Programme, Singapore General Hospital, Singapore
| | - Pratik Choudhary
- Leicester Diabetes Center, University of Leicester, Leicester, United Kingdom
| | | | - Yong Mong Bee
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Su-Yen Goh
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Ming Ming Teh
- Department of Endocrinology, Singapore General Hospital, Singapore
| |
Collapse
|
15
|
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F, Lamas S. Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med 2024; 222:85-105. [PMID: 38838921 DOI: 10.1016/j.freeradbiomed.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.
Collapse
Affiliation(s)
- Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Fernando Rodríguez-Pascual
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
16
|
Girardi ACC, Polidoro JZ, Castro PC, Pio-Abreu A, Noronha IL, Drager LF. Mechanisms of heart failure and chronic kidney disease protection by SGLT2 inhibitors in nondiabetic conditions. Am J Physiol Cell Physiol 2024; 327:C525-C544. [PMID: 38881421 DOI: 10.1152/ajpcell.00143.2024] [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: 02/29/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2is), initially developed for type 2 diabetes (T2D) treatment, have demonstrated significant cardiovascular and renal benefits in heart failure (HF) and chronic kidney disease (CKD), irrespective of T2D. This review provides an analysis of the multifaceted mechanisms underlying the cardiorenal benefits of SGLT2i in HF and CKD outside of the T2D context. Eight major aspects of the protective effects of SGLT2i beyond glycemic control are explored: 1) the impact on renal hemodynamics and tubuloglomerular feedback; 2) the natriuretic effects via proximal tubule Na+/H+ exchanger NHE3 inhibition; 3) the modulation of neurohumoral pathways with evidence of attenuated sympathetic activity; 4) the impact on erythropoiesis, not only in the context of local hypoxia but also systemic inflammation and iron regulation; 5) the uricosuria and mitigation of the hyperuricemic environment in cardiorenal syndromes; 6) the multiorgan metabolic reprogramming including the potential induction of a fasting-like state, improvement in glucose and insulin tolerance, and stimulation of lipolysis and ketogenesis; 7) the vascular endothelial growth factor A (VEGF-A) upregulation and angiogenesis, and 8) the direct cardiac effects. The intricate interplay between renal, neurohumoral, metabolic, and cardiac effects underscores the complexity of SGLT2i actions and provides valuable insights into their therapeutic implications for HF and CKD. Furthermore, this review sets the stage for future research to evaluate the individual contributions of these mechanisms in diverse clinical settings.
Collapse
Affiliation(s)
- Adriana C C Girardi
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Juliano Z Polidoro
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo C Castro
- Laboratório de Genética e Cardiologia Molecular, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Andrea Pio-Abreu
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Irene L Noronha
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luciano F Drager
- Disciplina de Nefrologia, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
- Unidade de Hipertensão, Faculdade de Medicina, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Marumo T, Yoshida N, Inoue N, Yamanouchi M, Ubara Y, Urakami S, Fujii T, Takazawa Y, Ohashi K, Kawarazaki W, Nishimoto M, Ayuzawa N, Hirohama D, Nagae G, Fujimoto M, Arai E, Kanai Y, Hoshino J, Fujita T. Aberrant proximal tubule DNA methylation underlies phenotypic changes related to kidney dysfunction in patients with diabetes. Am J Physiol Renal Physiol 2024; 327:F397-F411. [PMID: 38961842 DOI: 10.1152/ajprenal.00124.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Epigenetic mechanisms are considered to contribute to diabetic nephropathy by maintaining memory of poor glycemic control during the early stages of diabetes. However, DNA methylation changes in the human kidney are poorly characterized, because of the lack of cell type-specific analysis. We examined DNA methylation in proximal tubules (PTs) purified from patients with diabetic nephropathy and identified differentially methylated CpG sites, given the critical role of proximal tubules in the kidney injury. Hypermethylation was observed at CpG sites annotated to genes responsible for proximal tubule functions, including gluconeogenesis, nicotinamide adenine dinucleotide synthesis, transporters of glucose, water, phosphate, and drugs, in diabetic kidneys, whereas genes involved in oxidative stress and the cytoskeleton exhibited demethylation. Methylation levels of CpG sites annotated to ACTN1, BCAR1, MYH9, UBE4B, AFMID, TRAF2, TXNIP, FOXO3, and HNF4A were correlated with the estimated glomerular filtration rate, whereas methylation of the CpG site in RUNX1 was associated with interstitial fibrosis and tubular atrophy. Hypermethylation of G6PC and HNF4A was accompanied by decreased expression in diabetic kidneys. Proximal tubule-specific hypomethylation of metabolic genes related to HNF4A observed in control kidneys was compromised in diabetic kidneys, suggesting a role for aberrant DNA methylation in the dedifferentiation process. Multiple genes with aberrant DNA methylation in diabetes overlapped genes with altered expressions in maladaptive proximal tubule cells, including transcription factors PPARA and RREB1. In conclusion, DNA methylation derangement in the proximal tubules of patients with diabetes may drive phenotypic changes, characterized by inflammatory and fibrotic features, along with impaired function in metabolism and transport.NEW & NOTEWORTHY Cell type-specific DNA methylation patterns in the human kidney are not known. We examined DNA methylation in proximal tubules of patients with diabetic nephropathy and revealed that oxidative stress, cytoskeleton, and metabolism genes were aberrantly methylated. The results indicate that aberrant DNA methylation in proximal tubules underlies kidney dysfunction in diabetic nephropathy. Aberrant methylation could be a target for reversing memory of poor glycemic control.
Collapse
Affiliation(s)
- Takeshi Marumo
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Naoto Yoshida
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Noriko Inoue
- Nephrology Center, Toranomon Hospital, Tokyo, Japan
| | | | | | | | - Takeshi Fujii
- Department of Pathology, Toranomon Hospital, Tokyo, Japan
| | | | - Kenichi Ohashi
- Department of Human Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wakako Kawarazaki
- Department of Pharmacology, School of Medicine, International University of Health and Welfare, Chiba, Japan
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Mitsuhiro Nishimoto
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Ayuzawa
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Daigoro Hirohama
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Genta Nagae
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Mao Fujimoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Junichi Hoshino
- Nephrology Center, Toranomon Hospital, Tokyo, Japan
- Deparment of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Rinaldi A, Cippà PE, Nemazanyy I, Anglicheau D, Pallet N. Taurine Deficiency Is a Hallmark of Injured Kidney Allografts. Transplantation 2024; 108:e218-e228. [PMID: 39167563 DOI: 10.1097/tp.0000000000004987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
BACKGROUND Taurine is one of the most abundant amino acids in humans. Low taurine levels are associated with cellular senescence, mitochondrial dysfunction, DNA damage, and inflammation in mouse, all of which can be reversed by supplementation. It is unknown whether taurine metabolism is associated with kidney allograft function and survival. METHODS We performed urine metabolomic profiling of kidney transplant recipients in the early and late phases after transplantation combined with transcriptomic analysis of human kidney allografts. Single-nucleus RNA sequencing data sets of mouse kidneys after ischemia-reperfusion injury were analyzed. We analyzed the association of urinary taurine levels and taurine metabolism genes with kidney function, histology, and graft survival. RESULTS Urine taurine concentrations were significantly lower in kidney transplant recipients who experienced delayed graft function. In a mouse model of ischemia-reperfusion injury, the taurine biosynthesis gene, CSAD , but not the taurine transporter SLC6A6 , was repressed. In the late stage of transplantation, low level of taurine in urine was associated with impaired kidney function and chronic structural changes. Urine taurine level in the lowest tertile was predictive of graft loss. Expression of the taurine transporter SLC6A6 in the upper median, but not CSAD , was associated with chronic kidney injury and was predictive of graft loss. CONCLUSIONS Low urine taurine level is a marker of injury in the kidney allograft, is associated with poor kidney function, is associated with chronic histological changes, and is predictive of graft survival. The differential expression of CSAD and SLC6A6 , depending on the time after transplantation and marks of injury, highlights different mechanisms affecting taurine metabolism.
Collapse
Affiliation(s)
- Anna Rinaldi
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Pietro E Cippà
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Institut National de la Santé et de la Recherche Médicale (INSERM) US24/CNRS UMS3633, Paris, France
| | - Dany Anglicheau
- INSERM U1151, Université Paris Cité, Paris, France
- Service de Néphrologie et Transplantation, Assistance Publique Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Nicolas Pallet
- Service de Biochimie, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Université de Paris, INSERM UMRS1138, Centre de Recherche des Cordeliers, Paris, France
- Service de Néphrologie, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
19
|
Huang J, Hao J, Wang P, Xu Y. The Role of Mitochondrial Dysfunction in CKD-Related Vascular Calcification: From Mechanisms to Therapeutics. Kidney Int Rep 2024; 9:2596-2607. [PMID: 39291213 PMCID: PMC11403042 DOI: 10.1016/j.ekir.2024.05.005] [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: 02/27/2024] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 09/19/2024] Open
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD) and is closely associated with cardiovascular events. The transdifferentiation of vascular smooth muscles (VSMCs) into an osteogenic phenotype is hypothesized to be the primary cause underlying VC. However, there is currently no effective clinical treatment for VC. Growing evidence suggests that mitochondrial dysfunction accelerates the osteogenic differentiation of VSMCs and VC via multiple mechanisms. Therefore, elucidating the relationship between the osteogenic differentiation of VSMCs and mitochondrial dysfunction may assist in improving VC-related adverse clinical outcomes in patients with CKD. This review aimed to summarize the role of mitochondrial biogenesis, mitochondrial dynamics, mitophagy, and metabolic reprogramming, as well as mitochondria-associated oxidative stress (OS) and senescence in VC in patients with CKD to offer valuable insights into the clinical treatment of VC.
Collapse
Affiliation(s)
- Junmin Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfeng Hao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Peng Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yongzhi Xu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
20
|
Luo D, Lu X, Li H, Li Y, Wang Y, Jiang S, Li G, Xu Y, Wu K, Dou X, Liu Q, Chen W, Zhou Y, Mao H. The Spermine Oxidase/Spermine Axis Coordinates ATG5-Mediated Autophagy to Orchestrate Renal Senescence and Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306912. [PMID: 38775007 PMCID: PMC11304251 DOI: 10.1002/advs.202306912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/09/2024] [Indexed: 08/09/2024]
Abstract
Decreased plasma spermine levels are associated with kidney dysfunction. However, the role of spermine in kidney disease remains largely unknown. Herein, it is demonstrated that spermine oxidase (SMOX), a key enzyme governing polyamine metabolism, is predominantly induced in tubular epithelium of human and mouse fibrotic kidneys, alongside a reduction in renal spermine content in mice. Moreover, renal SMOX expression is positively correlated with kidney fibrosis and function decline in patients with chronic kidney disease. Importantly, supplementation with exogenous spermine or genetically deficient SMOX markedly improves autophagy, reduces senescence, and attenuates fibrosis in mouse kidneys. Further, downregulation of ATG5, a critical component of autophagy, in tubular epithelial cells enhances SMOX expression and reduces spermine in TGF-β1-induced fibrogenesis in vitro and kidney fibrosis in vivo. Mechanically, ATG5 readily interacts with SMOX under physiological conditions and in TGF-β1-induced fibrogenic responses to preserve cellular spermine levels. Collectively, the findings suggest SMOX/spermine axis is a potential novel therapy to antagonize renal fibrosis, possibly by coordinating autophagy and suppressing senescence.
Collapse
Affiliation(s)
- Dan Luo
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
- Department of NephrologyShunde HospitalSouthern Medical University (The First People's Hospital of Shunde)FoshanGuangdong528308China
| | - Xiaohui Lu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Hongyu Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yi Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yating Wang
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Simin Jiang
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Guanglan Li
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yiping Xu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Kefei Wu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Xianrui Dou
- Department of NephrologyShunde HospitalSouthern Medical University (The First People's Hospital of Shunde)FoshanGuangdong528308China
| | - Qinghua Liu
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Wei Chen
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Yi Zhou
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| | - Haiping Mao
- Department of NephrologyThe First Affiliated HospitalSun Yat‐sen UniversityNHC Key Laboratory of Clinical NephrologyGuangdong Provincial Key Laboratory of NephrologyGuangzhouGuangdong510080China
| |
Collapse
|
21
|
Huynh MT, Erfani Z, Al Nemri S, Chirayil S, Kovacs Z, Park JM. Enhanced Solubility and Polarization of 13C-Fumarate with Meglumine Allows for In Vivo Detection of Gluconeogenic Metabolism in Kidneys. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37435-37444. [PMID: 38984763 PMCID: PMC11272437 DOI: 10.1021/acsami.4c03163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Hyperpolarized 13C-labeled fumarate probes tissue necrosis via the production of 13C-malate. Despite its promises in detecting tumor necrosis and kidney injuries, its clinical translation has been limited, primarily due to the low solubility in conventional glassing solvents. In this study, we introduce a new formulation of fumarate for dissolution dynamic nuclear polarization (DNP) by using meglumine as a counterion, a nonmetabolizable derivative of sorbitol. We have found that meglumine fumarate vitrifies by itself with enhanced water solubility (4.8 M), which is expected to overcome the solubility-restricted maximum concentration of hyperpolarized fumarate after dissolution. The achievable liquid-state polarization level of meglumine-fumarate is more than doubled (29.4 ± 1.3%) as compared to conventional dimethyl sulfoxide (DMSO)-mixed fumarate (13.5 ± 2.4%). In vivo comparison of DMSO- and meglumine-prepared 50-mM hyperpolarized [1,4-13C2]fumarate shows that the signal sensitivity in rat kidneys increases by 10-fold. As a result, [1,4-13C2]aspartate and [13C]bicarbonate in addition to [1,4-13C2]malate can be detected in healthy rat kidneys in vivo using hyperpolarized meglumine [1,4-13C2]fumarate. In particular, the appearance of [13C]bicarbonate indicates that hyperpolarized meglumine [1,4-13C2]fumarate can be used to investigate phosphoenolpyruvate carboxykinase, a key regulatory enzyme in gluconeogenesis.
Collapse
Affiliation(s)
- Mai T Huynh
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Zohreh Erfani
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Sarah Al Nemri
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Sara Chirayil
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Zoltan Kovacs
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Jae Mo Park
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
22
|
Chowdhury TA, Mukuba D, Casabar M, Byrne C, Yaqoob MM. Management of diabetes in people with advanced chronic kidney disease. Diabet Med 2024:e15402. [PMID: 38992927 DOI: 10.1111/dme.15402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Diabetes is the commonest cause of end stage kidney disease globally, accounting for almost 40% of new cases requiring renal replacement therapy. Management of diabetes in people with advanced kidney disease on renal replacement therapy is challenging due to some unique aspects of assessment and treatment in this group of patients. Standard glycaemic assessment using glycated haemoglobin may not be valid in such patients due to altered red blood cell turnover or iron/erythropoietin deficiency, leading to changed red blood cell longevity. Therefore, use of continuous glucose monitoring may be beneficial to enable more focussed glycaemic assessment and improved adjustment of therapy. People with advanced kidney disease may be at higher risk of hypoglycaemia due to a number of physiological mechanisms, and in addition, therapeutic options are limited in such patients due to lack of experience or license. Insulin therapy is the basis of treatment of people with diabetes with advanced kidney disease due to many other drugs classes being contraindicated. Targets for glycaemic control should be adjusted according to co-morbidity and frailty, and continuous glucose monitoring should be used in people on dialysis to ensure low risk of hypoglycaemia. Post-transplant diabetes is common amongst people undergoing solid organ transplantation and confers a greater risk of mortality and morbidity in kidney transplant recipients. It should be actively screened for and managed in the post-transplant setting.
Collapse
Affiliation(s)
| | - Dorcas Mukuba
- Department of Diabetes, The Royal London Hospital, London, UK
| | - Mahalia Casabar
- Department of Nephrology, The Royal London Hospital, London, UK
| | - Conor Byrne
- Department of Nephrology, The Royal London Hospital, London, UK
| | - M Magdi Yaqoob
- Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
23
|
Zechner C, Rhee EP. Phosphate sensing in health and disease. Curr Opin Nephrol Hypertens 2024; 33:361-367. [PMID: 38572729 DOI: 10.1097/mnh.0000000000000984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW Disruptions of phosphate homeostasis are associated with a multitude of diseases with insufficient treatments. Our knowledge regarding the mechanisms underlying metazoan phosphate homeostasis and sensing is limited. Here, we highlight four major advancements in this field during the last 12-18 months. RECENT FINDINGS First, kidney glycolysis senses filtered phosphate, which results in the release of glycerol 3-phosphate (G-3-P). Circulating G-3-P then stimulates synthesis of the phosphaturic hormone fibroblast growth factor 23 in bone. Second, the liver serves as a postprandial phosphate reservoir to limit serum phosphate excursions. It senses phosphate ingestion and triggers renal excretion of excess phosphate through a nerve-dependent mechanism. Third, phosphate-starvation in cells massively induces the phosphate transporters SLC20A1/PiT1 and SLC20A2/PiT2, implying direct involvement of cellular phosphate sensing. Under basal phosphate-replete conditions, PiT1 is produced but immediately destroyed, which suggests a novel mechanism for the regulation of PiT1 abundance. Fourth, Drosophila melanogaster intestinal cells contain novel organelles called PXo bodies that limit intracellular phosphate excursions. Phosphate starvation leads to PXo body dissolution, which triggers midgut proliferation. SUMMARY These studies have opened novel avenues to dissect the mechanisms that govern metazoan phosphate sensing and homeostasis with the potential to identify urgently needed therapeutic targets.
Collapse
Affiliation(s)
- Christoph Zechner
- Division of Endocrinology, Department of Internal Medicine; Department of Pharmacology; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Eugene P Rhee
- Nephrology Division and Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Hasegawa K, Tamaki M, Shibata E, Inagaki T, Minato M, Yamaguchi S, Shimizu I, Miyakami S, Tada M, Wakino S. Ability of NAD and Sirt1 to epigenetically suppress albuminuria. Clin Exp Nephrol 2024; 28:599-607. [PMID: 38587753 PMCID: PMC11190001 DOI: 10.1007/s10157-024-02502-w] [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: 02/06/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
The time for diabetic nephropathy (DN) to progress from mild to severe is long. Thus, methods to continuously repress DN are required to exert long-lasting effects mediated through epigenetic regulation. In this study, we demonstrated the ability of nicotinamide adenine dinucleotide (NAD) and its metabolites to reduce albuminuria through Sirt1- or Nampt-dependent epigenetic regulation. We previously reported that proximal tubular Sirt1 was lowered before glomerular Sirt1. Repressed glomerular Sirt1 was found to epigenetically elevate Claudin-1. In addition, we reported that proximal tubular Nampt deficiency epigenetically augmented TIMP-1 levels in Sirt6-mediated pathways, leading to type-IV collagen deposition and diabetic fibrosis. Altogether, we propose that the Sirt1/Claudin-1 axis may be crucial in the onset of albuminuria at the early stages of DN and that the Nampt/Sirt6/TIMP-1 axis promotes diabetic fibrosis in the middle to late stages of DN. Finally, administration of NMN, an NAD precursor, epigenetically potentiates the regression of the onset of DN to maintain Sirt1 and repress Claudin-1 in podocytes, suggesting the potential use of NAD metabolites as epigenetic medications for DN.
Collapse
Affiliation(s)
- Kazuhiro Hasegawa
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Masanori Tamaki
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Eriko Shibata
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Taizo Inagaki
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masanori Minato
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Sumiyo Yamaguchi
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Ikuko Shimizu
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Shinji Miyakami
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Miho Tada
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Shu Wakino
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
25
|
Hasenour CM, Banerjee DR, Young JD. Metabolic Fluxes in the Renal Cortex Are Dysregulated In Vivo in Response to High-Fat Diet. Diabetes 2024; 73:903-908. [PMID: 38502790 PMCID: PMC11109784 DOI: 10.2337/db23-0710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Diabetes and obesity are risk factors for kidney disease. Whereas renal glucose production increases in diabetes, recent data suggest that gluconeogenic and oxidative capacity decline in kidney disease. Thus, metabolic dysregulation caused by diet-induced insulin resistance may sensitize the kidney for a loss in function. Here, we examined how diet-induced insulin resistance disrupts mitochondrial metabolic fluxes in the renal cortex in vivo. C57BL/6J mice were rendered insulin resistant through high-fat (HF) feeding; anaplerotic, cataplerotic, and oxidative metabolic fluxes in the cortex were quantified through 13C-isotope tracing during a hyperinsulinemic-euglycemic clamp. As expected, HF-fed mice exhibited increased body weight, gluconeogenesis, and systemic insulin resistance compared with chow-fed mice. Relative to the citric acid cycle, HF feeding increased metabolic flux through pyruvate carboxylation (anaplerosis) and phosphoenolpyruvate carboxykinase (cataplerosis) and decreased flux through the pyruvate dehydrogenase complex in the cortex. Furthermore, the relative flux from nonpyruvate sources of acetyl-CoA profoundly increased in the cortex of HF-fed mice, correlating with a marker of oxidative stress. The data demonstrate that HF feeding spares pyruvate from dehydrogenation at the expense of increasing cataplerosis, which may underpin renal gluconeogenesis during insulin resistance; the results also support the hypothesis that dysregulated oxidative metabolism in the kidney contributes to metabolic disease. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Clinton M. Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Deveena R. Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Jamey D. Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
26
|
Wang X, Chang HC, Gu X, Han W, Mao S, Lu L, Jiang S, Ding H, Han S, Qu X, Bao Z. Renal lipid accumulation and aging linked to tubular cells injury via ANGPTL4. Mech Ageing Dev 2024; 219:111932. [PMID: 38580082 DOI: 10.1016/j.mad.2024.111932] [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: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Renal tubular epithelial cells are vulnerable to stress-induced damage, including excessive lipid accumulation and aging, with ANGPTL4 potentially playing a crucial bridging role between these factors. In this study, RNA-sequencing was used to identify a marked increase in ANGPTL4 expression in kidneys of diet-induced obese and aging mice. Overexpression and knockout of ANGPTL4 in renal tubular epithelial cells (HK-2) was used to investigate the underlying mechanism. Subsequently, ANGPTL4 expression in plasma and kidney tissues of normal young controls and elderly individuals was analyzed using ELISA and immunohistochemical techniques. RNA sequencing results showed that ANGPTL4 expression was significantly upregulated in the kidney tissue of diet-induced obesity and aging mice. In vitro experiments demonstrated that overexpression of ANGPTL4 in HK-2 cells led to increased lipid deposition and senescence. Conversely, the absence of ANGPTL4 appears to alleviate the impact of free fatty acids (FFA) on aging in HK-2 cells. Additionally, aging HK-2 cells exhibited elevated ANGPTL4 expression, and stress response markers associated with cell cycle arrest. Furthermore, our clinical evidence revealed dysregulation of ANGPTL4 expression in serum and kidney tissue samples obtained from elderly individuals compared to young subjects. Our study findings indicate a potential association between ANGPTL4 and age-related metabolic disorders, as well as injury to renal tubular epithelial cells. This suggests that targeting ANGPTL4 could be a viable strategy for the clinical treatment of renal aging.
Collapse
Affiliation(s)
- Xiaojun Wang
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Hung-Chen Chang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Xuchao Gu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Wanlin Han
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Shihang Mao
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, China
| | - Lili Lu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Traditional Chinese Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Shuai Jiang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Thoracic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Haiyong Ding
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Urologic Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Shisheng Han
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xinkai Qu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Department of Cardiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| |
Collapse
|
27
|
Li H, Ren Q, Shi M, Ma L, Fu P. Lactate metabolism and acute kidney injury. Chin Med J (Engl) 2024:00029330-990000000-01083. [PMID: 38802283 DOI: 10.1097/cm9.0000000000003142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Indexed: 05/29/2024] Open
Abstract
ABSTRACT Acute kidney injury (AKI) is a common clinically critical syndrome in hospitalized patients with high morbidity and mortality. At present, the mechanism of AKI has not been fully elucidated, and no therapeutic drugs exist. As known, glycolytic product lactate is a key metabolite in physiological and pathological processes. The kidney is an important gluconeogenic organ, where lactate is the primary substrate of renal gluconeogenesis in physiological conditions. During AKI, altered glycolysis and gluconeogenesis in kidneys significantly disturb the lactate metabolic balance, which exert impacts on the severity and prognosis of AKI. Additionally, lactate-derived posttranslational modification, namely lactylation, is novel to AKI as it could regulate gene transcription of metabolic enzymes involved in glycolysis or Warburg effect. Protein lactylation widely exists in human tissues and may severely affect non-histone functions. Moreover, the strategies of intervening lactate metabolic pathways are expected to bring a new dawn for the treatment of AKI. This review focused on renal lactate metabolism, especially in proximal renal tubules after AKI, and updated recent advances of lactylation modification, which may help to explore potential therapeutic targets against AKI.
Collapse
Affiliation(s)
- Hui Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | |
Collapse
|
28
|
Trebuian CI, Marza AM, Chioibaş R, Şutoi D, Petrica A, Crintea-Najette I, Popa D, Borcan F, Flondor D, Mederle OA. Lactate Profile Assessment-A Good Predictor of Prognosis in Patients with COVID-19 and Septic Shock Requiring Continuous Renal Therapy. Clin Pract 2024; 14:980-994. [PMID: 38921256 PMCID: PMC11202829 DOI: 10.3390/clinpract14030078] [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: 04/17/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
INTRODUCTION Lactate is a useful prognostic marker, as its level increases in hypoxic tissue and/or during accelerated aerobic glycolysis due to excessive beta-adrenergic stimulation and decreased lactate clearance. The Surviving Sepsis Campaign Bundle 2018 Update suggests premeasurement of lactate within 2-4 h so that physicians perform, assist, administer, and introduce lactate-guided resuscitation to reduce mortality due to sepsis. METHODS A total of 108 patients with septic shock who underwent continuous renal replacement therapy (CRRT) for acute kidney injury were enrolled in this observational study. Demographic, clinical, and laboratory data were collected, and patients were divided into two groups: survivors and non-survivors. RESULTS Multivariate analysis demonstrated that lactate levels at 24 h after initiation of CRRT treatment, but not lactate levels at intensive care unit (ICU) admission, were associated with mortality. Lactate clearance was associated with lower mortality among the survivors (OR = 0.140) at 6 h after ICU admission and late mortality (OR = 0.260) after 24 h. The area under the ROC curves for mortality was 0.682 for initial lactate; 0.797 for lactate at 24 h; and 0.816 for lactate clearance at 24 h. CONCLUSIONS Our result reinforces that the determination of lactate dynamics represents a good predictor for mortality, and serial lactate measurements may be more useful prognostic markers than initial lactate in patients with septic shock.
Collapse
Affiliation(s)
- Cosmin Iosif Trebuian
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
- Department of Anesthesia and Intensive Care, Emergency County Hospital Resita, 320210 Resita, Romania
| | - Adina Maria Marza
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
- Emergency Department, Emergency Clinical Municipal Hospital Timisoara, 300079 Timisoara, Romania
| | - Raul Chioibaş
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
| | - Dumitru Şutoi
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
| | - Alina Petrica
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
- Emergency Department of “Pius Brinzeu”, Emergency Clinical County Hospital Timisoara, 300736 Timisoara, Romania
| | - Iulia Crintea-Najette
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
- Emergency Department, Emergency Clinical Municipal Hospital Timisoara, 300079 Timisoara, Romania
| | - Daian Popa
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
- Emergency Department, Emergency Clinical Municipal Hospital Timisoara, 300079 Timisoara, Romania
| | - Florin Borcan
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (F.B.); (D.F.)
| | - Daniela Flondor
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (F.B.); (D.F.)
- Research Center for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Ovidiu Alexandru Mederle
- Department of Surgery I, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (C.I.T.); (A.M.M.); (D.Ş.); (A.P.); (I.C.-N.); (D.P.); (O.A.M.)
| |
Collapse
|
29
|
Kuhn C, Mohebbi N, Ritter A. Metabolic acidosis in chronic kidney disease: mere consequence or also culprit? Pflugers Arch 2024; 476:579-592. [PMID: 38279993 PMCID: PMC11006741 DOI: 10.1007/s00424-024-02912-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/29/2024]
Abstract
Metabolic acidosis is a frequent complication in non-transplant chronic kidney disease (CKD) and after kidney transplantation. It occurs when net endogenous acid production exceeds net acid excretion. While nephron loss with reduced ammoniagenesis is the main cause of acid retention in non-transplant CKD patients, additional pathophysiological mechanisms are likely inflicted in kidney transplant recipients. Functional tubular damage by calcineurin inhibitors seems to play a key role causing renal tubular acidosis. Notably, experimental and clinical studies over the past decades have provided evidence that metabolic acidosis may not only be a consequence of CKD but also a driver of disease. In metabolic acidosis, activation of hormonal systems and the complement system resulting in fibrosis have been described. Further studies of changes in renal metabolism will likely contribute to a deeper understanding of the pathophysiology of metabolic acidosis in CKD. While alkali supplementation in case of reduced serum bicarbonate < 22 mmol/l has been endorsed by CKD guidelines for many years to slow renal functional decline, among other considerations, beneficial effects and thresholds for treatment have lately been under intense debate. This review article discusses this topic in light of the most recent results of trials assessing the efficacy of dietary and pharmacological interventions in CKD and kidney transplant patients.
Collapse
Affiliation(s)
- Christian Kuhn
- Clinic for Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | | | - Alexander Ritter
- Clinic for Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland.
- Clinic for Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Chrysopoulou M, Rinschen MM. Metabolic Rewiring and Communication: An Integrative View of Kidney Proximal Tubule Function. Annu Rev Physiol 2024; 86:405-427. [PMID: 38012048 DOI: 10.1146/annurev-physiol-042222-024724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The kidney proximal tubule is a key organ for human metabolism. The kidney responds to stress with altered metabolite transformation and perturbed metabolic pathways, an ultimate cause for kidney disease. Here, we review the proximal tubule's metabolic function through an integrative view of transport, metabolism, and function, and embed it in the context of metabolome-wide data-driven research. Function (filtration, transport, secretion, and reabsorption), metabolite transformation, and metabolite signaling determine kidney metabolic rewiring in disease. Energy metabolism and substrates for key metabolic pathways are orchestrated by metabolite sensors. Given the importance of renal function for the inner milieu, we also review metabolic communication routes with other organs. Exciting research opportunities exist to understand metabolic perturbation of kidney and proximal tubule function, for example, in hypertension-associated kidney disease. We argue that, based on the integrative view outlined here, kidney diseases without genetic cause should be approached scientifically as metabolic diseases.
Collapse
Affiliation(s)
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark;
- III. Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
31
|
Rauckhorst AJ, Vasquez Martinez G, Mayoral Andrade G, Wen H, Kim JY, Simoni A, Robles-Planells C, Mapuskar KA, Rastogi P, Steinbach EJ, McCormick ML, Allen BG, Pabla NS, Jackson AR, Coleman MC, Spitz DR, Taylor EB, Zepeda-Orozco D. Tubular mitochondrial pyruvate carrier disruption elicits redox adaptations that protect from acute kidney injury. Mol Metab 2024; 79:101849. [PMID: 38056691 PMCID: PMC10733108 DOI: 10.1016/j.molmet.2023.101849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
OBJECTIVE Energy-intensive kidney reabsorption processes essential for normal whole-body function are maintained by tubular epithelial cell metabolism. Although tubular metabolism changes markedly following acute kidney injury (AKI), it remains unclear which metabolic alterations are beneficial or detrimental. By analyzing large-scale, publicly available datasets, we observed that AKI consistently leads to downregulation of the mitochondrial pyruvate carrier (MPC). This investigation aimed to understand the contribution of the tubular MPC to kidney function, metabolism, and acute injury severity. METHODS We generated tubular epithelial cell-specific Mpc1 knockout (MPC TubKO) mice and employed renal function tests, in vivo renal 13C-glucose tracing, mechanistic enzyme activity assays, and tests of injury and survival in an established rhabdomyolysis model of AKI. RESULTS MPC TubKO mice retained normal kidney function, displayed unchanged markers of kidney injury, but exhibited coordinately increased enzyme activities of the pentose phosphate pathway and the glutathione and thioredoxin oxidant defense systems. Following rhabdomyolysis-induced AKI, compared to WT control mice, MPC TubKO mice showed increased glycolysis, decreased kidney injury and oxidative stress markers, and strikingly increased survival. CONCLUSIONS Our findings suggest that decreased renal tubular mitochondrial pyruvate uptake hormetically upregulates oxidant defense systems before AKI and is a beneficial adaptive response after rhabdomyolysis-induced AKI. This raises the possibility of therapeutically modulating the MPC to attenuate AKI severity.
Collapse
Affiliation(s)
- Adam J Rauckhorst
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa, Iowa City, IA, USA
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA
| | - Gabriel Mayoral Andrade
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA
| | - Hsiang Wen
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Aaron Simoni
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA
| | - Claudia Robles-Planells
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA
| | - Kranti A Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Prerna Rastogi
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Emily J Steinbach
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ashley R Jackson
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Mitchell C Coleman
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA; Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Eric B Taylor
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA, USA; FOEDRC Metabolomics Core Research Facility, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA; Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA.
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA; Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Faivre A, Dissard R, Kuo W, Verissimo T, Legouis D, Arnoux G, Heckenmeyer C, Fernandez M, Tihy M, Rajaram RD, Delitsikou V, Le NA, Spingler B, Mueller B, Shulz G, Lindenmeyer M, Cohen C, Rutkowski JM, Moll S, Scholz CC, Kurtcuoglu V, de Seigneux S. Evolution of hypoxia and hypoxia-inducible factor asparaginyl hydroxylase regulation in chronic kidney disease. Nephrol Dial Transplant 2023; 38:2276-2288. [PMID: 37096392 PMCID: PMC10539236 DOI: 10.1093/ndt/gfad075] [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: 06/03/2022] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND The roles of hypoxia and hypoxia inducible factor (HIF) during chronic kidney disease (CKD) are much debated. Interventional studies with HIF-α activation in rodents have yielded contradictory results. The HIF pathway is regulated by prolyl and asparaginyl hydroxylases. While prolyl hydroxylase inhibition is a well-known method to stabilize HIF-α, little is known about the effect asparaginyl hydroxylase factor inhibiting HIF (FIH). METHODS We used a model of progressive proteinuric CKD and a model of obstructive nephropathy with unilateral fibrosis. In these models we assessed hypoxia with pimonidazole and vascularization with three-dimensional micro-computed tomography imaging. We analysed a database of 217 CKD biopsies from stage 1 to 5 and we randomly collected 15 CKD biopsies of various severity degrees to assess FIH expression. Finally, we modulated FIH activity in vitro and in vivo using a pharmacologic approach to assess its relevance in CKD. RESULTS In our model of proteinuric CKD, we show that early CKD stages are not characterized by hypoxia or HIF activation. At late CKD stages, some areas of hypoxia are observed, but these are not colocalizing with fibrosis. In mice and in humans, we observed a downregulation of the HIF pathway, together with an increased FIH expression in CKD, according to its severity. Modulating FIH in vitro affects cellular metabolism, as described previously. In vivo, pharmacologic FIH inhibition increases the glomerular filtration rate of control and CKD animals and is associated with decreased development of fibrosis. CONCLUSIONS The causative role of hypoxia and HIF activation in CKD progression is questioned. A pharmacological approach of FIH downregulation seems promising in proteinuric kidney disease.
Collapse
Affiliation(s)
- Anna Faivre
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Romain Dissard
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Willy Kuo
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- National Centre of Competence in Research, Kidney. CH, University of Zurich, Zurich, Switzerland
| | - Thomas Verissimo
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - David Legouis
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Grégoire Arnoux
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Clinical Pathology, Department of Pathology and Immunology, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Carolyn Heckenmeyer
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Marylise Fernandez
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Matthieu Tihy
- Service of Clinical Pathology, Department of Pathology and Immunology, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Renuga D Rajaram
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Vasiliki Delitsikou
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Ngoc An Le
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | | | - Bert Mueller
- Biomaterials Science Center, Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Georg Shulz
- Biomaterials Science Center, Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
- Micro- and Nanotomography Core Facility, Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Maja Lindenmeyer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clemens Cohen
- Nephrological Center, Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany
| | - Joseph M Rutkowski
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Solange Moll
- Service of Clinical Pathology, Department of Pathology and Immunology, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Carsten C Scholz
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- National Centre of Competence in Research, Kidney. CH, University of Zurich, Zurich, Switzerland
- Institute of Physiology, University Medicine Greifswald, Greifswald, Germany
| | - Vartan Kurtcuoglu
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- National Centre of Competence in Research, Kidney. CH, University of Zurich, Zurich, Switzerland
| | - Sophie de Seigneux
- Department of Medicine and Cell physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Service of Nephrology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
- National Centre of Competence in Research, Kidney. CH, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Parvathareddy VP, Wu J, Thomas SS. Insulin Resistance and Insulin Handling in Chronic Kidney Disease. Compr Physiol 2023; 13:5069-5076. [PMID: 37770191 PMCID: PMC11079812 DOI: 10.1002/cphy.c220019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Insulin regulates energy metabolism involving multiple organ systems. Insulin resistance (IR) occurs when organs exhibit reduced insulin sensitivity, leading to difficulties in maintaining glucose homeostasis. IR ensures decades prior to development of overt diabetes and can cause silent metabolic derangements. IR is typically seen very early in the course of chronic kidney disease (CKD) and is evident even when the estimated glomerular filtration rate (eGFR) is within the normal range and IR persists at various stages of kidney disease. In this article, we will discuss insulin handling by the kidneys, mechanisms responsible for IR in CKD, measurements and management of IR in patients with CKD, and recent type 2 diabetic trials with implications for improved cardiovascular outcomes in CKD. © 2023 American Physiological Society. Compr Physiol 13:5069-5076, 2023.
Collapse
Affiliation(s)
- Vishnu P. Parvathareddy
- Nephrology Division, Department of Medicine, Baylor
College of Medicine, Houston, Texas, USA
| | - Jiao Wu
- Nephrology Division, Department of Medicine, Baylor
College of Medicine, Houston, Texas, USA
| | - Sandhya S. Thomas
- Nephrology Division, Department of Medicine, Michael E.
Debakey VA Medical Center, Houston, Texas, USA
- Nephrology Division, Department of Medicine, Baylor
College of Medicine, Houston, Texas, USA
| |
Collapse
|
34
|
Packer M. Fetal Reprogramming of Nutrient Surplus Signaling, O-GlcNAcylation, and the Evolution of CKD. J Am Soc Nephrol 2023; 34:1480-1491. [PMID: 37340541 PMCID: PMC10482065 DOI: 10.1681/asn.0000000000000177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
ABSTRACT Fetal kidney development is characterized by increased uptake of glucose, ATP production by glycolysis, and upregulation of mammalian target of rapamycin (mTOR) and hypoxia-inducible factor-1 alpha (HIF-1 α ), which (acting in concert) promote nephrogenesis in a hypoxic low-tubular-workload environment. By contrast, the healthy adult kidney is characterized by upregulation of sirtuin-1 and adenosine monophosphate-activated protein kinase, which enhances ATP production through fatty acid oxidation to fulfill the needs of a normoxic high-tubular-workload environment. During stress or injury, the kidney reverts to a fetal signaling program, which is adaptive in the short term, but is deleterious if sustained for prolonged periods when both oxygen tension and tubular workload are heightened. Prolonged increases in glucose uptake in glomerular and proximal tubular cells lead to enhanced flux through the hexosamine biosynthesis pathway; its end product-uridine diphosphate N -acetylglucosamine-drives the rapid and reversible O-GlcNAcylation of thousands of intracellular proteins, typically those that are not membrane-bound or secreted. Both O-GlcNAcylation and phosphorylation act at serine/threonine residues, but whereas phosphorylation is regulated by hundreds of specific kinases and phosphatases, O-GlcNAcylation is regulated only by O-GlcNAc transferase and O-GlcNAcase, which adds or removes N-acetylglucosamine, respectively, from target proteins. Diabetic and nondiabetic CKD is characterized by fetal reprogramming (with upregulation of mTOR and HIF-1 α ) and increased O-GlcNAcylation, both experimentally and clinically. Augmentation of O-GlcNAcylation in the adult kidney enhances oxidative stress, cell cycle entry, apoptosis, and activation of proinflammatory and profibrotic pathways, and it inhibits megalin-mediated albumin endocytosis in glomerular mesangial and proximal tubular cells-effects that can be aggravated and attenuated by augmentation and muting of O-GlcNAcylation, respectively. In addition, drugs with known nephroprotective effects-angiotensin receptor blockers, mineralocorticoid receptor antagonists, and sodium-glucose cotransporter 2 inhibitors-are accompanied by diminished O-GlcNAcylation in the kidney, although the role of such suppression in mediating their benefits has not been explored. The available evidence supports further work on the role of uridine diphosphate N -acetylglucosamine as a critical nutrient surplus sensor (acting in concert with upregulated mTOR and HIF-1 α signaling) in the development of diabetic and nondiabetic CKD.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute , Dallas , Texas and Imperial College , London , United Kingdom
| |
Collapse
|
35
|
Nistor M, Schmidt M, Klingner C, Klingner C, Schwab M, Bischoff SJ, Matziolis G, Rodríguez-González GL, Schiffner R. Renal Glucose Release after Unilateral Renal Denervation during a Hypoglycemic Clamp in Pigs with an Altered Hypothalamic Pituitary Adrenal Axis after Late-Gestational Dexamethasone Injection. Int J Mol Sci 2023; 24:12738. [PMID: 37628918 PMCID: PMC10454812 DOI: 10.3390/ijms241612738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Previously, we demonstrated in pigs that renal denervation halves glucose release during hypoglycaemia and that a prenatal dexamethasone injection caused increased ACTH and cortisol concentrations as markers of a heightened hypothalamic pituitary adrenal axis (HPAA) during hypoglycaemia. In this study, we investigated the influence of an altered HPAA on renal glucose release during hypoglycaemia. Pigs whose mothers had received two late-gestational dexamethasone injections were subjected to a 75 min hyperinsulinaemic-hypoglycaemic clamp (<3 mmol/L) after unilateral surgical denervation. Para-aminohippurate (PAH) clearance, inulin, sodium excretion and arterio-venous blood glucose difference were measured every fifteen minutes. The statistical analysis was performed with a Wilcoxon signed-rank test. PAH, inulin, the calculated glomerular filtration rate and plasma flow did not change through renal denervation. Urinary sodium excretion increased significantly (p = 0.019). Side-dependent renal net glucose release (SGN) decreased by 25 ± 23% (p = 0.004). At 25 percent, the SGN decrease was only half of that observed in non-HPAA-altered animals in our prior investigation. The current findings may suggest that specimens with an elevated HPAA undergo long-term adaptations to maintain glucose homeostasis. Nonetheless, the decrease in SGN warrants further investigations and potentially caution in performing renal denervation in certain patient groups, such as diabetics at risk of hypoglycaemia.
Collapse
Affiliation(s)
- Marius Nistor
- Orthopaedic Department, Jena University Hospital, Campus Eisenberg, 07607 Eisenberg, Germany; (M.N.)
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, 07743 Jena, Germany
| | - Carsten Klingner
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany (M.S.)
| | - Caroline Klingner
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany (M.S.)
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany (M.S.)
| | | | - Georg Matziolis
- Orthopaedic Department, Jena University Hospital, Campus Eisenberg, 07607 Eisenberg, Germany; (M.N.)
| | | | - René Schiffner
- Orthopaedic Department, Jena University Hospital, Campus Eisenberg, 07607 Eisenberg, Germany; (M.N.)
- Emergency Department, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Emergency Department, Helios University Clinic Wuppertal, 42283 Wuppertal, Germany
| |
Collapse
|
36
|
Saade MC, Parikh SM. Energy Metabolism in CKD: Running Low on Fuel. KIDNEY360 2023; 4:1014-1016. [PMID: 37651663 PMCID: PMC10484351 DOI: 10.34067/kid.0000000000000231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 07/10/2023] [Indexed: 09/02/2023]
Affiliation(s)
- Marie Christelle Saade
- Division of Nephrology , Department of Medicine , University of Texas Southwestern , Dallas , Texas
| | | |
Collapse
|
37
|
Dalga D, Verissimo T, de Seigneux S. Gluconeogenesis in the kidney: in health and in chronic kidney disease. Clin Kidney J 2023; 16:1249-1257. [PMID: 37529654 PMCID: PMC10387387 DOI: 10.1093/ckj/sfad046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Indexed: 08/03/2023] Open
Abstract
Chronic kidney disease (CKD) is a global health issue with increasing prevalence. Despite large improvements in current therapies, slowing CKD progression remains a challenge. A better understanding of renal pathophysiology is needed to offer new therapeutic targets. The role of metabolism alterations and mitochondrial dysfunction in tubular cells is increasingly recognized in CKD progression. In proximal tubular cells, CKD progression is associated with a switch from fatty acid oxidation to glycolysis. Glucose synthesis through gluconeogenesis is one of the principal physiological functions of the kidney. Loss of tubular gluconeogenesis in a stage-dependent manner is a key feature of CKD and contributes to systemic and possibly local metabolic complications. The local consequences observed may be related to an accumulation of precursors, such as glycogen, but also to the various physiological functions of the gluconeogenesis enzymes. The basic features of metabolism in proximal tubular cells and their modifications during CKD will be reviewed. The metabolic modifications and their influence on kidney disease will be described, as well as the local and systemic consequences. Finally, therapeutic interventions will be discussed.
Collapse
Affiliation(s)
- Delal Dalga
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Thomas Verissimo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
38
|
Miguel V, Kramann R. Metabolic reprogramming heterogeneity in chronic kidney disease. FEBS Open Bio 2023; 13:1154-1163. [PMID: 36723270 PMCID: PMC10315765 DOI: 10.1002/2211-5463.13568] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/02/2023] Open
Abstract
Fibrosis driven by excessive accumulation of extracellular matrix (ECM) is the hallmark of chronic kidney disease (CKD). Myofibroblasts, which are the cells responsible for ECM production, are activated by cross talk with injured proximal tubule and immune cells. Emerging evidence suggests that alterations in metabolism are not only a feature of but also play an influential role in the pathogenesis of renal fibrosis. The application of omics technologies to cell-tracing animal models and follow-up functional data suggest that cell-type-specific metabolic shifts have particular roles in the fibrogenic response. In this review, we cover the main metabolic reprogramming outcomes in renal fibrosis and provide a future perspective on the field of renal fibrometabolism.
Collapse
Affiliation(s)
- Verónica Miguel
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University HospitalAachenGermany
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University HospitalAachenGermany
| |
Collapse
|
39
|
Cippà PE, McMahon AP. Proximal tubule responses to injury: interrogation by single-cell transcriptomics. Curr Opin Nephrol Hypertens 2023; 32:352-358. [PMID: 37074682 PMCID: PMC10330172 DOI: 10.1097/mnh.0000000000000893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) occurs in approximately 10-15% of patients admitted to hospital and is associated with adverse clinical outcomes. Despite recent advances, management of patients with AKI is still mainly supportive, including the avoidance of nephrotoxins, volume and haemodynamic management and renal replacement therapy. A better understanding of the renal response to injury is the prerequisite to overcome current limitations in AKI diagnostics and therapy. RECENT FINDINGS Single-cell technologies provided new opportunities to study the complexity of the kidney and have been instrumental for rapid advancements in the understanding of the cellular and molecular mechanisms of AKI. SUMMARY We provide an update on single-cell technologies and we summarize the recent discoveries on the cellular response to injury in proximal tubule cells from the early response in AKI, to the mechanisms of tubule repair and the relevance of maladaptive tubule repair in the transition to chronic kidney disease.
Collapse
Affiliation(s)
- Pietro E Cippà
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculity of Biomedical Sciences, Università della Svizzera Italiana, Lugano Switzerland
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Verissimo T, Dalga D, Arnoux G, Sakhi I, Faivre A, Auwerx H, Bourgeois S, Paolucci D, Gex Q, Rutkowski JM, Legouis D, Wagner CA, Hall AM, de Seigneux S. PCK1 is a key regulator of metabolic and mitochondrial functions in renal tubular cells. Am J Physiol Renal Physiol 2023; 324:F532-F543. [PMID: 37102687 PMCID: PMC10202477 DOI: 10.1152/ajprenal.00038.2023] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
Phosphoenolpyruvate carboxykinase 1 (PCK1 or PEPCK-C) is a cytosolic enzyme converting oxaloacetate to phosphoenolpyruvate, with a potential role in gluconeogenesis, ammoniagenesis, and cataplerosis in the liver. Kidney proximal tubule cells display high expression of this enzyme, whose importance is currently not well defined. We generated PCK1 kidney-specific knockout and knockin mice under the tubular cell-specific PAX8 promoter. We studied the effect of PCK1 deletion and overexpression at the renal level on tubular physiology under normal conditions and during metabolic acidosis and proteinuric renal disease. PCK1 deletion led to hyperchloremic metabolic acidosis characterized by reduced but not abolished ammoniagenesis. PCK1 deletion also resulted in glycosuria, lactaturia, and altered systemic glucose and lactate metabolism at baseline and during metabolic acidosis. Metabolic acidosis resulted in kidney injury in PCK1-deficient animals with decreased creatinine clearance and albuminuria. PCK1 further regulated energy production by the proximal tubule, and PCK1 deletion decreased ATP generation. In proteinuric chronic kidney disease, mitigation of PCK1 downregulation led to better renal function preservation. PCK1 is essential for kidney tubular cell acid-base control, mitochondrial function, and glucose/lactate homeostasis. Loss of PCK1 increases tubular injury during acidosis. Mitigating kidney tubular PCK1 downregulation during proteinuric renal disease improves renal function.NEW & NOTEWORTHY Phosphoenolpyruvate carboxykinase 1 (PCK1) is highly expressed in the proximal tubule. We show here that this enzyme is crucial for the maintenance of normal tubular physiology, lactate, and glucose homeostasis. PCK1 is a regulator of acid-base balance and ammoniagenesis. Preventing PCK1 downregulation during renal injury improves renal function, rendering it an important target during renal disease.
Collapse
Affiliation(s)
- Thomas Verissimo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| | - Delal Dalga
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| | - Grégoire Arnoux
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Imene Sakhi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Anna Faivre
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Hannah Auwerx
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Deborah Paolucci
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Quentin Gex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | | - David Legouis
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Sophie de Seigneux
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
41
|
Aouad H, Faucher Q, Sauvage FL, Pinault E, Barrot CC, Arnion H, Essig M, Marquet P. A multi-omics investigation of tacrolimus off-target effects on a proximal tubule cell-line. Pharmacol Res 2023; 192:106794. [PMID: 37187266 DOI: 10.1016/j.phrs.2023.106794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Tacrolimus, an immunosuppressive drug prescribed to a majority of organ transplant recipients is nephrotoxic, through still unclear mechanisms. This study on a lineage of proximal tubular cells using a multi-omics approach aims to detect off-target pathways modulated by tacrolimus that can explain its nephrotoxicity. METHODS LLC-PK1 cells were exposed to 5µM of tacrolimus for 24h in order to saturate its therapeutic target FKBP12 and other high-affine FKBPs and favour its binding to less affine targets. Intracellular proteins and metabolites, and extracellular metabolites were extracted and analysed by LC-MS/MS. The transcriptional expression of the dysregulated proteins PCK-1, as well as of the other gluconeogenesis-limiting enzymes FBP1 and FBP2, was measured using RT-qPCR. Cell viability with this concentration of tacrolimus was further checked until 72h. RESULTS In our cell model of acute exposure to a high concentration of tacrolimus, different metabolic pathways were impacted including those of arginine (e.g., citrulline, ornithine) (p<0.0001), amino acids (e.g., valine, isoleucine, aspartic acid) (p<0.0001) and pyrimidine (p<0.01). In addition, it induced oxidative stress (p<0.01) as shown by a decrease in total cell glutathione quantity. It impacted cell energy through an increase in Krebs cycle intermediates (e.g., citrate, aconitate, fumarate) (p<0.01) and down-regulation of PCK-1 (p<0.05) and FPB1 (p<0.01), which are key enzymes in gluconeogenesis and acid-base balance control. DISCUSSION The variations found using a multi-omics pharmacological approach clearly point towards a dysregulation of energy production and decreased gluconeogenesis, a hallmark of chronic kidney disease which may also be an important toxicity pathways of tacrolimus.
Collapse
Affiliation(s)
- Hassan Aouad
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France
| | - Quentin Faucher
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France
| | | | - Emilie Pinault
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France
| | - Claire-Cécile Barrot
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France
| | - Hélène Arnion
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France
| | - Marie Essig
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France; Department of Nephrology, CHU Limoges, Limoges, France
| | - Pierre Marquet
- Pharmacology & Transplantation, Université de Limoges, INSERM U1248, Limoges, France; Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France.
| |
Collapse
|
42
|
Wen L, Wei Q, Livingston MJ, Dong G, Li S, Hu X, Li Y, Huo Y, Dong Z. PFKFB3 mediates tubular cell death in cisplatin nephrotoxicity by activating CDK4. Transl Res 2023; 253:31-40. [PMID: 36243313 PMCID: PMC10416729 DOI: 10.1016/j.trsl.2022.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Nephrotoxicity is a major side effect of cisplatin, a widely used cancer therapy drug. However, the mechanism of cisplatin nephrotoxicity remains unclear and no effective kidney protective strategies are available. Here, we report the induction of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in both in vitro cell culture and in vivo mouse models of cisplatin nephrotoxicity. Notably, PFKFB3 was mainly induced in the nucleus of kidney tubular cells, suggesting a novel function other than its canonical role in glycolysis. Both pharmacological inhibition and genetic silencing of PFKFB3 led to the suppression of cisplatin-induced apoptosis in cultured renal proximal tubular cells (RPTCs). Moreover, cisplatin-induced kidney injury or nephrotoxicity was ameliorated in renal proximal tubule-specific PFKFB3 knockout mice. Mechanistically, we demonstrated the interaction of PFKFB3 with cyclin-dependent kinase 4 (CDK4) during cisplatin treatment, resulting in CDK4 activation and consequent phosphorylation and inactivation of retinoblastoma tumor suppressor (Rb). Inhibition of CDK4 reduced cisplatin-induced apoptosis in RPTCs and kidney injury in mice. Collectively, this study unveils a novel pathological role of PFKFB3 in cisplatin nephrotoxicity through the activation of the CDK4/Rb pathway, suggesting a new kidney protective strategy for cancer patients by blocking PFKFB3.
Collapse
Affiliation(s)
- Lu Wen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Siyao Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Xiaoru Hu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ying Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
43
|
The mechanisms of alkali therapy in targeting renal diseases. Biochem Soc Trans 2023; 51:223-232. [PMID: 36744634 DOI: 10.1042/bst20220690] [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: 11/08/2022] [Revised: 12/28/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is characterized by progressive reduction in kidney function and treatments aiming at stabilizing or slowing its progression may avoid or delay the necessity of kidney replacement therapy and the increased mortality associated with reduced kidney function. Metabolic acidosis, and less severe stages of the acid stress continuum, are common consequences of CKD and some interventional studies support that its correction slows the progression to end-stage kidney disease. This correction can be achieved with mineral alkali in the form of bicarbonate or citrate salts, ingestion of diets with fewer acid-producing food components or more base-producing food components, or a pharmacological approach. In this mini-review article, we summarize the potential mechanisms involved in the beneficial effects of alkali therapy. We also discuss the perspectives in the field and challenges that must be overcome to advance our understanding of such mechanisms.
Collapse
|
44
|
Rauckhorst AJ, Martinez GV, Andrade GM, Wen H, Kim JY, Simoni A, Mapuskar KA, Rastogi P, Steinbach EJ, McCormick ML, Allen BG, Pabla NS, Jackson AR, Coleman MC, Spitz DR, Taylor EB, Zepeda-Orozco D. Tubular Mitochondrial Pyruvate Carrier Disruption Elicits Redox Adaptations that Protect from Acute Kidney Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526492. [PMID: 36778297 PMCID: PMC9915694 DOI: 10.1101/2023.01.31.526492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Energy-intensive kidney reabsorption processes essential for normal whole-body function are maintained by tubular epithelial cell metabolism. Tubular metabolism changes markedly following acute kidney injury (AKI), but which changes are adaptive versus maladaptive remain poorly understood. In publicly available data sets, we noticed a consistent downregulation of the mitochondrial pyruvate carrier (MPC) after AKI, which we experimentally confirmed. To test the functional consequences of MPC downregulation, we generated novel tubular epithelial cell-specific Mpc1 knockout (MPC TubKO) mice. 13C-glucose tracing, steady-state metabolomic profiling, and enzymatic activity assays revealed that MPC TubKO coordinately increased activities of the pentose phosphate pathway and the glutathione and thioredoxin oxidant defense systems. Following rhabdomyolysis-induced AKI, MPC TubKO decreased markers of kidney injury and oxidative damage and strikingly increased survival. Our findings suggest that decreased mitochondrial pyruvate uptake is a central adaptive response following AKI and raise the possibility of therapeutically modulating the MPC to attenuate AKI severity.
Collapse
Affiliation(s)
- Adam J. Rauckhorst
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA, USA
- FOEDRC Metabolomics Core Research Facility, University of Iowa, Iowa City, IA, USA
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH, USA
| | - Gabriel Mayoral Andrade
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH, USA
| | - Hsiang Wen
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Aaron Simoni
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH, USA
| | - Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Prerna Rastogi
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Emily J Steinbach
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Michael L. McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Navjot S. Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ashley R. Jackson
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Mitchell C. Coleman
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Eric B. Taylor
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA, USA
- FOEDRC Metabolomics Core Research Facility, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus OH, USA
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
45
|
Phase Angle Association with Dietary Habits and Metabolic Syndrome in Diabetic Hypertensive Patients: A Cross-Sectional Study. Nutrients 2022; 14:nu14235058. [PMID: 36501088 PMCID: PMC9738996 DOI: 10.3390/nu14235058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Phase angle (PhA) levels are often lower than normal because both disease-specific parameters and disease-related inflammatory status, metabolic syndrome (MetS) included, can affect PhA. Therefore, the aim of this cross-sectional study was to compare body composition, metabolic profile and dietary patterns of participants with arterial hypertension (AH), type 2 diabetes mellitus (T2DM) and MetS with regard to PhA values. A total of 208 participants were included, of whom 53.6% were males. For each participant, data about body composition and anthropometric parameters, clinical and laboratory parameters, as well as food frequency questionnaire (FFQ) and Mediterranean Diet Serving Score (MDSS) were obtained. MC-780 Multi Frequency Segmental Body Mass Analyzer (Tanita) was used to assess body composition. Furthermore, waist-to-hip ratio (WHR) and waist-to-height ratio (WHtR) were calculated. The results showed that 75 (36.06%) participants had low PhA values and 133 (63.94%) had high PhA values. Participants with higher PhA values had significantly higher body fat percentage (p = 0.04), fat-free mass (kg; p < 0.001), muscle mass (kg; p < 0.001), skeletal muscle mass (% and kg; p < 0.001), sarcopenic index (SMI; p < 0.001) and mid-upper arm circumference (MUAC; p = 0.04), as well as lower fat mass percentage (p = 0.04). Regarding food frequency consumption, significantly higher intakes of red meat (p = 0.003), poultry (p = 0.02) and fast food (p = 0.003) were noticed in participants with higher PhA values. Adherence to the Mediterranean Diet (MeDi) was exceptionally low in both groups of participants, with significantly higher fish intake noticed in participants with high PhA (p = 0.03). In conclusion, our results showed that body composition could be the indicator of PhA in MetS as well as overall low adherence to the MeDi principles. These findings highlight the importance of adequate nutritional strategies and novel approaches to maintaining optimal body composition and adopting proper eating habits within the framework of one’s disease.
Collapse
|
46
|
Rinaldi A, Lazareth H, Poindessous V, Nemazanyy I, Sampaio JL, Malpetti D, Bignon Y, Naesens M, Rabant M, Anglicheau D, Cippà PE, Pallet N. Impaired fatty acid metabolism perpetuates lipotoxicity along the transition to chronic kidney injury. JCI Insight 2022; 7:161783. [PMID: 35998043 DOI: 10.1172/jci.insight.161783] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Energy metabolism failure in proximal tubule cells (PTC) is a hallmark of chronic kidney injury. We combined transcriptomic, metabolomic and lipidomic approaches in experimental models and patient cohorts to investigate the molecular bases of the progression to chronic kidney allograft injury initiated by ischemia-reperfusion injury (IRI). The urinary metabolome of kidney transplant recipients with chronic allograft injury and who experienced severe IRI was significantly enriched with long chain fatty acids (FA). We identified a renal FA-related gene signature with low levels of Cpt2 and Acsm5 and high levels of Acsl4 and Acsm5 associated with IRI, transition to chronic injury, and established CKD in mouse models and kidney transplant recipients. The findings were consistent with the presence of Cpt2-, Acsl4+, Acsl5+, Acsm5- PTC failing to recover from IRI as identified by snRNAseq. In vitro experiments indicated that endoplasmic reticulum (ER) stress contributes to CPT2 repression, which, in turn, promotes lipids accumulation, drives profibrogenic epithelial phenotypic changes, and activates the unfolded protein response. ER stress through CPT2 inhibition and lipid accumulation, engages an auto-amplification loop leading to lipotoxicity and self-sustained cellular stress. Thus, IRI imprints a persistent FA metabolism disturbance in the proximal tubule sustaining the progression to chronic kidney allograft injury.
Collapse
Affiliation(s)
- Anna Rinaldi
- Department of Medicine, Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Hélène Lazareth
- Centre de Recherche des Cordeliers, INSERM U1138, Paris, France
| | | | - Ivan Nemazanyy
- PMM: The Metabolism-Metabolome Platform, Necker Federative Research Structu, INSERM US24/CNRS, UMS3633, Paris, France
| | - Julio L Sampaio
- CurieCoreTech Metabolomics and Lipidomics Technology Platform, Paris, France
| | - Daniele Malpetti
- Instituto Dalle Molle di Studi sull'Intelligenza Artificiale, Lugano, Switzerland
| | - Yohan Bignon
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Marion Rabant
- Department of Pathology, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Dany Anglicheau
- Department of Kidney Transplantation, Necker Hospital, Paris, France
| | - Pietro E Cippà
- Department of Medicine, Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Nicolas Pallet
- Centre de Recherche des Cordeliers, INSERM U1138, Paris, France
| |
Collapse
|
47
|
Legouis D, Criton G, Assouline B, Le Terrier C, Sgardello S, Pugin J, Marchi E, Sangla F. Unsupervised clustering reveals phenotypes of AKI in ICU COVID-19 patients. Front Med (Lausanne) 2022; 9:980160. [PMID: 36275817 PMCID: PMC9579431 DOI: 10.3389/fmed.2022.980160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Acute Kidney Injury (AKI) is a very frequent condition, occurring in about one in three patients admitted to an intensive care unit (ICU). AKI is a syndrome defined as a sudden decrease in glomerular filtration rate. However, this unified definition does not reflect the various mechanisms involved in AKI pathophysiology, each with its own characteristics and sensitivity to therapy. In this study, we aimed at developing an innovative machine learning based method able to subphenotype AKI according to its pattern of risk factors. Methods We adopted a three-step pipeline of analyses. First, we looked for factors associated with AKI using a generalized additive model. Second, we calculated the importance of each identified AKI related factor in the estimated AKI risk to find the main risk factor for AKI, at the single patient level. Lastly, we clusterized AKI patients according to their profile of risk factors and compared the clinical characteristics and outcome of every cluster. We applied this method to a cohort of severe COVID-19 patients hospitalized in the ICU of the Geneva University Hospitals. Results Among the 248 patients analyzed, we found 7 factors associated with AKI development. Using the individual expression of these factors, we identified three groups of AKI patients, based on the use of Lopinavir/Ritonavir, baseline eGFR, use of dexamethasone and AKI severity. The three clusters expressed distinct characteristics in terms of AKI severity and recovery, metabolic patterns and hospital mortality. Conclusion We propose here a new method to phenotype AKI patients according to their most important individual risk factors for AKI development. When applied to an ICU cohort of COVID-19 patients, we were able to differentiate three groups of patients. Each expressed specific AKI characteristics and outcomes, which probably reflect a distinct pathophysiology.
Collapse
Affiliation(s)
- David Legouis
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
- Laboratory of Nephrology, Department of Medicine and Cell Physiology, University Hospital of Geneva, Geneva, Switzerland
- *Correspondence: David Legouis
| | - Gilles Criton
- Geneva School of Economics and Management, University of Geneva, Geneva, Switzerland
| | - Benjamin Assouline
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Christophe Le Terrier
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Sebastian Sgardello
- Department of Surgery, Center Hospitalier du Valais Romand, Sion, Switzerland
| | - Jérôme Pugin
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Elisa Marchi
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Frédéric Sangla
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
48
|
Guo L, Chen S, Ou L, Li S, Ye ZN, Liu HF. Disrupted Alpha-Ketoglutarate Homeostasis: Understanding Kidney Diseases from the View of Metabolism and Beyond. Diabetes Metab Syndr Obes 2022; 15:1961-1974. [PMID: 35783031 PMCID: PMC9248815 DOI: 10.2147/dmso.s369090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/17/2022] [Indexed: 11/26/2022] Open
Abstract
Alpha-ketoglutarate (AKG) is a key intermediate of various metabolic pathways including tricarboxylic acid (TCA) cycle, anabolic and catabolic reactions of amino acids, and collagen biosynthesis. Meanwhile, AKG also participates in multiple signaling pathways related to cellular redox regulation, epigenetic processes, and inflammation response. Emerging evidence has shown that kidney diseases like diabetic nephropathy and renal ischemia/reperfusion injury are associated with metabolic disorders. In consistence with metabolic role of AKG, further metabolomics study demonstrated a dysregulated AKG level in kidney diseases. Intriguingly, earlier studies during the years of 1980s and 1990s indicated that AKG may benefit wound healing and surgery recovery. Recently, interests on AKG are arising again due to its protective roles on healthy ageing, which may shed light on developing novel therapeutic strategies against age-related diseases including renal diseases. This review will summarize the physiological and pathological properties of AKG, as well as the underlying molecular mechanisms, with a special emphasis on kidney diseases.
Collapse
Affiliation(s)
- Lijing Guo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Shihua Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Liping Ou
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Shangmei Li
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Zhen-Nan Ye
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Correspondence: Zhen-Nan Ye; Hua-Feng Liu, Email ;
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| |
Collapse
|