1
|
Wang J, Chen J, Li Z, Liu Z. The negative feedback loop of NF-κB/miR-202-5p/HMGB2 attenuates sepsis induced acute kidney injury. Int Immunopharmacol 2024; 142:113050. [PMID: 39226829 DOI: 10.1016/j.intimp.2024.113050] [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: 07/12/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
Sepsis represents a primary cause of acute kidney injury (AKI), yet the underlying mechanisms of septic AKI remain poorly understood. Thus, there exists an urgent need for a deeper understanding of its underlying mechanisms and the development of effective therapeutic strategies. Our study reveals a notable induction in microRNA-202-5p (miR-202-5p) levels within renal tubular cells in septic AKI both in vivo and in vitro models. Treatment of renal tubular cells with LPS induced NF-κB activation, which was linked to the induction of miR-202-5p. ChIP assays confirmed NF-κB binding to the miR-202-5p gene promoter upon LPS stimulation. Functionally, miR-202-5p mimics attenuated tubular cell death, kidney injury, and intra-renal inflammatory cytokine production, whereas inhibition of miR-202-5p conferred injurious effects in septic AKI. Notably, miR-202-5p suppressed the expression of High Mobility Group Box 2 (HMGB2) in both in vitro and in vivo septic AKI models. Luciferase microRNA target assays further validated HMGB2 as a direct target of miR-202-5p. Knockdown of HMGB2 inhibits LPS-induced NF-κB activation in septic AKI, as evidenced by HMGB2 siRNA transfection significantly inhibited the nuclear translocation of NF-κB. Together, these findings elucidate the NF-κB/miR-202-5p/HMGB2 negative feedback loop which can attenuate kidney injury by inhibiting renal inflammation in septic AKI. Our findings open new avenues for developing targeted therapies to manage septic AKI effectively.
Collapse
Affiliation(s)
- Juan Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jian Chen
- qi ting jie dao she qu wei sheng fu wu zhong xin, Yixing, Jiangsu, China
| | - Zheng Li
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| |
Collapse
|
2
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [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: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
3
|
Yan M, Zhang N, Quan L, Bin W, Xi J, Dou C, Liu Z, Gui Y, Yin LH. NF-κB/miR-455-5p/SOCS3 Axis Aggravates Sepsis-Induced Acute Kidney Injury through Promoting Renal Inflammation. Nephron Clin Pract 2024:1-12. [PMID: 39378861 DOI: 10.1159/000541727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Sepsis is the leading contributor to acute kidney injury (AKI), responsible for 45-70% of AKI occurrences. Despite this, septic AKI is a highly multifactorial and complex condition, and our grasp of its pathogenesis is still not fully developed. Consequently, there remains a significant gap in effective diagnostic and therapeutic strategies for septic AKI. METHODS In the in vitro experiments, BUMPT cells were exposed to lipopolysaccharides (LPS). In vivo experiments involved inducing sepsis in mice through administration of LPS injections. Additionally, in certain experiments, either a miR-455-5p mimic or an anti-miR-455-5p LAN was administered to the mice via injections into the tail vein. The mice were then sacrificed 24 h following LPS administration for subsequent analysis. RESULTS We observed a significant elevation in miR-455-5p levels within renal tubular cells following LPS-induced septic AKI. Our investigation revealed that NF-κB plays a crucial role in the upregulation of miR-455-5p. Inhibition of NF-κB using TPCA-1 prevented the rise in miR-455-5p levels in BUMPT cells (mouse proximal tubular cells from Boston University) cultured in vitro. Chromatin immunoprecipitation assays confirmed that NF-κB directly interacts with the promoter region of the miR-455-5p gene in response to LPS treatment. Functionally, introducing miR-455-5p mimics intensified cell apoptosis, kidney damage, and the production of inflammatory cytokines, while silencing miR-455-5p had protective effects in septic mice. Notably, administering anti-miR-455-5p enhanced SOCS3 expression, whereas miR-455-5p mimics reduced SOCS3 levels following LPS exposure. Furthermore, our luciferase reporter assays demonstrated that SOCS3 is a direct target of miR-455-5p. CONCLUSION This study indicates an NF-κB/miR-455-5p/SOCS3 axis which can exacerbate kidney damage by enhancing renal inflammation. This process highlights potential therapeutic targets for managing septic AKI.
Collapse
Affiliation(s)
- Mingjuan Yan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China,
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China,
| | - Ni Zhang
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Li Quan
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Wei Bin
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Jing Xi
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Caoshuai Dou
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Yongfeng Gui
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changsha, China
| | - Liang-Hong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Tokumaru K, Imafuku T, Satoh T, Inazumi T, Hirashima S, Nishinoiri A, Nagasaki T, Maeda H, Sugimoto Y, Tanaka M, Matsushita K, Maruyama T, Watanabe H. Omega 3 Fatty Acids Attenuate the Acute Kidney Injury to CKD Transition and Renal Fibrosis: Identification of Antifibrotic Metabolites. KIDNEY360 2024; 5:1422-1434. [PMID: 39259608 DOI: 10.34067/kid.0000000574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Key Points
Omega-3 polyunsaturated fatty acids prevent AKI to CKD transition and renal fibrosis.Eicosapentaenoic acid metabolites 18-hydroxyeicosapentaenoic acid, 17,18-epoxyeicosatetraenoic acid, and 17,18-dihydroxyeicosatetraenoic acid have antifibrotic effects.
Background
AKI is an established risk factor for developing CKD. Recently, the renoprotective effect of omega-3 polyunsaturated fatty acids (ω3PUFAs) has attracted attention. The aims of this study were to evaluate the effect of ω3PUFAs on the transition of AKI to CKD and to identify fatty acid active metabolites in renal tissue.
Methods
Two mice models of the AKI to CKD transition (7-week, male) and unilateral ureteral obstruction–induced renal fibrosis (11-week, male) were fed linseed oil, rich in ω3PUFAs (Lin group), or with soybean oil, low in ω3PUFAs (Soy group). Renal fatty acids and metabolites composition in mice were measured by liquid chromatography-mass spectrometry. Rat renal fibroblast cells were used for in vitro study.
Results
At day 14 after 35 minutes of bilateral renal ischemia reperfusion, significant increase in survival was observed in the Lin group compared with the Soy group. Using the 30-minute bilateral renal ischemia–reperfusion model (AKI to CKD model), the Lin group showed attenuated renal tissue damage and fibrosis. In addition, the antifibrotic effect of the Lin group was also observed in the unilateral ureteral obstruction renal fibrosis model. In the two mice models, levels of eicosapentaenoic acid (EPA) and its metabolites were significantly elevated in renal tissue of mice fed with Lin. Cultured NRK-49F incubated with EPA and its metabolites 18-hydroxyeicosapentaenoic acid, 17,18-epoxyeicosatetraenoic acid, and 17,18-dihydroxyeicosatetraenoic acid displayed suppressed TGF-β1–stimulated α-smooth muscle actin protein expression. These effects were suppressed in the presence of an inhibitor of a cytochrome P450 involved in EPA metabolism. This observation suggests that the EPA metabolites have antifibrotic effects.
Conclusions
ω3PUFAs prevent the AKI to CKD transition and renal fibrosis. Moreover, the EPA metabolites 18-hydroxyeicosapentaenoic acid, 17,18-epoxyeicosatetraenoic acid, and 17,18-dihydroxyeicosatetraenoic acid were found to have antifibrotic effects.
Collapse
Affiliation(s)
- Kai Tokumaru
- Department of Clinical Pharmacy and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadashi Imafuku
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takao Satoh
- Kumamoto Industrial Research Institute, Kumamoto, Japan
| | - Tomoaki Inazumi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shu Hirashima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ayano Nishinoiri
- Department of Clinical Pharmacy and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Motoko Tanaka
- Department of Nephrology, Akebono Clinic, Kumamoto, Japan
| | | | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Watanabe
- Department of Clinical Pharmacy and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
5
|
Maeoka Y, Bradford T, Su XT, Sharma A, Yang CL, Ellison DH, McCormick JA, Cornelius RJ. Distal convoluted tubule-specific disruption of the COP9 signalosome but not its regulatory target cullin 3 causes tubular injury. Am J Physiol Renal Physiol 2024; 327:F667-F682. [PMID: 39205661 PMCID: PMC11483082 DOI: 10.1152/ajprenal.00138.2024] [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: 05/06/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
The disease familial hyperkalemic hypertension (FHHt; also known as Gordon syndrome) is caused by aberrant accumulation of with-no-lysine kinase (WNK4) activating the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) of the kidney. Mutations in cullin 3 (CUL3) cause FHHt by disrupting interaction with the deneddylase COP9 signalosome (CSN). Deletion of Cul3 or Jab1 (the catalytically active CSN subunit) along the entire nephron causes a partial FHHt phenotype with activation of the WNK4-STE20/SPS1-related proline/alanine-rich kinase (SPAK)-NCC pathway. However, progressive kidney injury likely prevents hypertension, hyperkalemia, and hyperchloremic metabolic acidosis associated with FHHt. We hypothesized that DCT-specific deletion would more closely model the disease. We used Slc12a3-Cre-ERT2 mice to delete Cul3 (DCT-Cul3-/-) or Jab1 (DCT-Jab1-/-) only in the DCT and examined the mice after short- and long-term deletion. Short-term DCT-specific knockout of both Cul3 and Jab1 mice caused elevated WNK4, pSPAKS373, and pNCCT53 abundance. However, neither model demonstrated changes in plasma K+, Cl-, or total CO2, even though no injury was present. Long-term DCT-Jab1-/- mice showed significantly lower NCC and parvalbumin abundance and a higher abundance of kidney injury molecule-1, a marker of proximal tubule injury. No injury or reduction in NCC or parvalbumin was observed in long-term DCT-Cul3-/- mice. In summary, the prevention of injury outside the DCT did not lead to a complete FHHt phenotype despite activation of the WNK4-SPAK-NCC pathway, possibly due to insufficient NCC activation. Chronically, only DCT-Jab1-/- mice developed tubule injury and atrophy of the DCT, suggesting a direct JAB1 effect or dysregulation of other cullins as mechanisms for injury.NEW & NOTEWORTHY CUL3 degrades WNK4, which prevents activation of NCC in the DCT. CSN regulation of CUL3 is impaired in the disease FHHt, causing accumulation of WNK4. Short-term DCT-specific disruption of CUL3 or the CSN in mice resulted in activation of the WNK4-SPAK-NCC pathway but not hyperkalemic metabolic acidosis found in FHHt. Tubule injury was observed only after long-term CSN disruption. The data suggest that disruption of other cullins may be the cause for the injury.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Tanner Bradford
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- LeDucq Transatlantic Network of Excellence, Boston, Massachusetts, United States
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
6
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed Western Diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.592234. [PMID: 38798535 PMCID: PMC11118422 DOI: 10.1101/2024.05.11.592234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Pharmacological inhibition of megalin (also known as low-density lipoprotein receptor-related protein 2: LRP2) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. Methods Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and -/- littermates. To study atherosclerosis, all mice were bred to an LDL receptor -/- background and fed a Western diet to induce atherosclerosis. Results PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed the Western diet, but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished albumin accumulation in PTCs within 10 days of Western diet feeding. RNA sequencing analyses demonstrated the upregulation of inflammation-related pathways in kidney. Conclusions PTC-specific megalin deletion does not affect atherosclerosis, but leads to tubulointerstitial nephritis in mice fed Western diet, with severe pathologies in male mice.
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | | | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
7
|
Miao J, Zhu H, Wang J, Chen J, Han F, Lin W. Experimental models for preclinical research in kidney disease. Zool Res 2024; 45:1161-1174. [PMID: 39257378 PMCID: PMC11491777 DOI: 10.24272/j.issn.2095-8137.2024.072] [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: 03/03/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are significant public health issues associated with a long-term increase in mortality risk, resulting from various etiologies including renal ischemia, sepsis, drug toxicity, and diabetes mellitus. Numerous preclinical models have been developed to deepen our understanding of the pathophysiological mechanisms and therapeutic approaches for kidney diseases. Among these, rodent models have proven to be powerful tools in the discovery of novel therapeutics, while the development of kidney organoids has emerged as a promising advancement in the field. This review provides a comprehensive analysis of the construction methodologies, underlying biological mechanisms, and recent therapeutic developments across different AKI and CKD models. Additionally, this review summarizes the advantages, limitations, and challenges inherent in these preclinical models, thereby contributing robust evidence to support the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jin Miao
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Huanhuan Zhu
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Junni Wang
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Jianghua Chen
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Fei Han
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China. E-mail:
| | - Weiqiang Lin
- Department of Nephrology, Center for Regeneration and Aging Medicine, Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China. E-mail:
| |
Collapse
|
8
|
Fan Y, Dong S, Xia Y, Yang X, Lei Q, Xu F, Liang D, Liang S, Zhang M, Yang F, Jing Y, Li L, Zhu X, Bao H, Chen Z, Zeng C. Role of TSP-1 and its receptor ITGB3 in the renal tubulointerstitial injury of focal segmental glomerulosclerosis. J Biol Chem 2024; 300:107516. [PMID: 38960036 PMCID: PMC11339032 DOI: 10.1016/j.jbc.2024.107516] [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: 02/20/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 07/05/2024] Open
Abstract
Focal segmental glomerulosclerosis (FSGS), a common cause of primary glomerulonephritis, has a poor prognosis and is pathologically featured by tubulointerstitial injury. Thrombospondin-1 (TSP-1) is an extracellular matrix protein that acts in combination with different receptors in the kidney. Here, we analyzed the tubular expression of TSP-1 and its receptor integrin β3 (ITGB3) in FSGS. Previously the renal interstitial chip analysis of FSGS patients with tubular interstitial injury showed that the expression of TSP-1 and ITGB3 were upregulated. We found that the expression of TSP-1 and ITGB3 increased in the tubular cells of FSGS patients. The plasma level of TSP-1 increased and was correlated to the degree of tubulointerstitial lesions in FSGS patients. TSP-1/ITGB3 signaling induced renal tubular injury in HK-2 cells exposure to bovine serum albumin and the adriamycin (ADR)-induced nephropathy model. THBS1 KO ameliorated tubular injury and renal fibrosis in ADR-treated mice. THBS1 knockdown decreased the expression of KIM-1 and caspase 3 in the HK-2 cells treated with bovine serum albumin, while THBS1 overexpression could induce tubular injury. In vivo, we identified cyclo-RGDfK as an agent to block the binding of TSP-1 to ITGB3. Cyclo-RGDfK treatment could alleviate ADR-induced renal tubular injury and interstitial fibrosis in mice. Moreover, TSP-1 and ITGB3 were colocalized in tubular cells of FSGS patients and ADR-treated mice. Taken together, our data showed that TSP-1/ITGB3 signaling contributed to the development of renal tubulointerstitial injury in FSGS, potentially identifying a new therapeutic target for FSGS.
Collapse
Affiliation(s)
- Yun Fan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Medical University, Nanjing, China; the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Shihui Dong
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanyuan Xia
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xue Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qunjuan Lei
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Feng Xu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Dandan Liang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shaoshan Liang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fan Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Jing
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lijuan Li
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaodong Zhu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hao Bao
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaohong Chen
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing Medical University, Nanjing, China; National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Huang S, Lu H, Chen J, Jiang C, Jiang G, Maduraiveeran G, Pan Y, Liu J, Deng LE. Advances in drug delivery-based therapeutic strategies for renal fibrosis treatment. J Mater Chem B 2024; 12:6532-6549. [PMID: 38913013 DOI: 10.1039/d4tb00737a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Renal fibrosis is the result of all chronic kidney diseases and is becoming a major global health hazard. Currently, traditional treatments for renal fibrosis are difficult to meet clinical needs due to shortcomings such as poor efficacy or highly toxic side effects. Therefore, therapeutic strategies that target the kidneys are needed to overcome these shortcomings. Drug delivery can be attained by improving drug stability and addressing controlled release and targeted delivery of drugs in the delivery category. By combining drug delivery technology with nanosystems, controlled drug release and biodistribution can be achieved, enhancing therapeutic efficacy and reducing toxic cross-wise effects. This review discusses nanomaterial drug delivery strategies reported in recent years. Firstly, the present review describes the mechanisms of renal fibrosis and anti-renal fibrosis drug delivery. Secondly, different nanomaterial drug delivery strategies for the treatment of renal injury and fibrosis are highlighted. Finally, the limitations of these strategies are also discussed. Investigating various anti-renal fibrosis drug delivery strategies reveals the characteristics and therapeutic effects of various novel nanosystem-derived drug delivery approaches. This will serve as a reference for future research on drug delivery strategies for renal fibrosis treatment.
Collapse
Affiliation(s)
- Sida Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Hanqi Lu
- Department of Nephrology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China.
| | - Jin Chen
- Department of Nephrology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China.
| | - Chengyi Jiang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Guanmin Jiang
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan people's hospital), 78 Wandao Road South, Dongguan, 523059 Guangdong, China.
| | - Govindhan Maduraiveeran
- Materials Electrochemistry Laboratory, Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur - 603 203, Chengalpattu, Tamil Nadu, India.
| | - Ying Pan
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China.
| | - Li-Er Deng
- Department of Nephrology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China.
| |
Collapse
|
10
|
Ma R, Ouyang H, Meng S, Liu J, Tian J, Jia N, Liu Y, Xu X, Yang X, Hou FF. Urinary cytokeratin 20 as a predictor for chronic kidney disease following acute kidney injury. JCI Insight 2024; 9:e180326. [PMID: 38805402 PMCID: PMC11383368 DOI: 10.1172/jci.insight.180326] [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: 02/15/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUNDIdentifying patients with acute kidney injury (AKI) at high risk of chronic kidney disease (CKD) progression remains a challenge.METHODSKidney transcriptome sequencing was applied to identify the top upregulated genes in mice with AKI. The product of the top-ranking gene was identified in tubular cells and urine in mouse and human AKI. Two cohorts of patients with prehospitalization estimated glomerular filtration rate (eGFR) ≥ 45 mL/min/1.73 m2 who survived over 90 days after AKI were used to derive and validate the predictive models. AKI-CKD progression was defined as eGFR < 60 mL/min/1.73 m2 and with minimum 25% reduction from baseline 90 days after AKI in patients with prehospitalization eGFR ≥ 60 mL/min/1.73 m2. AKI-advanced CKD was defined as eGFR < 30 mL/min/1.73 m2 90 days after AKI in those with prehospitalization eGFR 45-59 mL/min/1.73 m2.RESULTSKidney cytokeratin 20 (CK20) was upregulated in injured proximal tubular cells and detectable in urine within 7 days after AKI. High concentrations of urinary CK20 (uCK20) were independently associated with the severity of histological AKI and the risk of AKI-CKD progression. In the Test set, the AUC of uCK20 for predicting AKI-CKD was 0.80, outperforming reported biomarkers for predicting AKI. Adding uCK20 to clinical variables improved the ability to predict AKI-CKD progression, with an AUC of 0.90, and improved the risk reclassification.CONCLUSIONThese findings highlight uCK20 as a useful predictor for AKI-CKD progression and may provide a tool to identify patients at high risk of CKD following AKI.FUNDINGNational Natural Science Foundation of China, National Key R&D Program of China, 111 Plan, Guangdong Key R&D Program.
Collapse
|
11
|
Möckel T, Boegel S, Schwarting A. Transcriptome Analysis of BAFF/BAFF-R System in Murine Nephrotoxic Serum Nephritis. Int J Mol Sci 2024; 25:5415. [PMID: 38791453 PMCID: PMC11121395 DOI: 10.3390/ijms25105415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is an emerging cause for morbidity and mortality worldwide. Acute kidney injury (AKI) can transition to CKD and finally to end-stage renal disease (ESRD). Targeted treatment is still unavailable. NF-κB signaling is associated with CKD and activated by B cell activating factor (BAFF) via BAFF-R binding. In turn, renal tubular epithelial cells (TECs) are critical for the progression of fibrosis and producing BAFF. Therefore, the direct involvement of the BAFF/BAFF-R system to the pathogenesis of CKD is conceivable. We performed non-accelerated nephrotoxic serum nephritis (NTN) as the CKD model in BAFF KO (B6.129S2-Tnfsf13btm1Msc/J), BAFF-R KO (B6(Cg)-Tnfrsf13ctm1Mass/J) and wildtype (C57BL/6J) mice to analyze the BAFF/BAFF-R system in anti-glomerular basement membrane (GBM) disease using high throughput RNA sequencing. We found that BAFF signaling is directly involved in the upregulation of collagen III as BAFF ko mice showed a reduced expression. However, these effects were not mediated via BAFF-R. We identified several upregulated genes that could explain the effects of BAFF in chronic kidney injury such as Txnip, Gpx3, Igfbp7, Ccn2, Kap, Umod and Ren1. Thus, we conclude that targeted treatment with anti-BAFF drugs such as belimumab may reduce chronic kidney damage. Furthermore, upregulated genes may be useful prognostic CKD biomarkers.
Collapse
Affiliation(s)
- Tamara Möckel
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
| | - Sebastian Boegel
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
| | - Andreas Schwarting
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (T.M.); (S.B.)
- Center for Rheumatic Disease Rhineland-Palatinate GmbH, 55543 Bad Kreuznach, Germany
| |
Collapse
|
12
|
Li ZL, Huang MM, Yu MY, Nie DF, Fu SL, Di JJ, Lan T, Liu BC, Wu QL. Mitochondrial fumarate promotes ischemia/reperfusion-induced tubular injury. Acta Physiol (Oxf) 2024; 240:e14121. [PMID: 38409944 DOI: 10.1111/apha.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
AIM Mitochondrial dysfunction, a characteristic pathological feature of renal Ischemic/reperfusion injury (I/RI), predisposes tubular epithelial cells to maintain an inflammatory microenvironment, however, the exact mechanisms through which mitochondrial dysfunction modulates the induction of tubular injury remains incompletely understood. METHODS ESI-QTRAP-MS/MS approach was used to characterize the targeted metabolic profiling of kidney with I/RI. Tubule injury, mitochondrial dysfunction, and fumarate level were evaluated using qPCR, transmission electron microscopy, ELISA, and immunohistochemistry. RESULTS We demonstrated that tubule injury occurred at the phase of reperfusion in murine model of I/RI. Meanwhile, enhanced glycolysis and mitochondrial dysfunction were found to be associated with tubule injury. Further, we found that tubular fumarate, which resulted from fumarate hydratase deficiency and released from dysfunctional mitochondria, promoted tubular injury. Mechanistically, fumarate induced tubular injury by causing disturbance of glutathione (GSH) hemostasis. Suppression of GSH with buthionine sulphoximine administration could deteriorate the fumarate inhibition-mediated tubule injury recovery. Reactive oxygen species/NF-κB signaling activation played a vital role in fumarate-mediated tubule injury. CONCLUSION Our studies demonstrated that the mitochondrial-derived fumarate promotes tubular epithelial cell injury in renal I/RI. Blockade of fumarate-mediated ROS/NF-κB signaling activation may serve as a novel therapeutic approach to ameliorate hypoxic tubule injury.
Collapse
Affiliation(s)
- Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Ming-Min Huang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Meng-Yao Yu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Di-Fei Nie
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Sha-Li Fu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jing-Jing Di
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Ting Lan
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Qiu-Li Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Ohata K, Sugaya T, Nguyen HN, Arai K, Hatanaka Y, Uno K, Tohma M, Uechi T, Sekiguchi K, Oikawa T, Nagabukuro H, Kuniyeda K, Kamijo-Ikemori A, Suzuki-Kemuriyama N, Nakae D, Noiri E, Miyajima K. Urinary liver-type fatty acid binding protein is a biomarker reflecting renal damage and the ameliorative effect of drugs at an early stage of histone-induced acute kidney injury. Nephrology (Carlton) 2024; 29:117-125. [PMID: 37950597 DOI: 10.1111/nep.14254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
AIM Circulated histones play a crucial role in the pathogenesis of infectious diseases and severe trauma, and it is one of the potential molecular targets for therapeutics. Recently, we reported that histone is one of the causative agents for urinary L-FABP increase. However, the mechanism is still unclear, especially in severe cases. We further investigated the mechanism of urinary L-FABP increase using a more severe mouse model with histone-induced kidney injury. This study also aims to evaluate the therapeutic responsiveness of urinary L-FABP as a preliminary study. METHODS Human L-FABP chromosomal transgenic mice were administrated 30 mg/kg histone from a tail vein with a single dose. We also performed a comparative study in LPS administration model. For the evaluation of the therapeutic responsiveness of urinary L-FABP, we used heparin and rolipram. RESULTS The histological change with cast formation as a characteristic of the models was observed in proximal tubules. Urinary L-FABP levels were significantly elevated and these levels tended to be higher in those with more cast formation. Heparin and rolipram had the ameliorative effect of the cast formation induced by histone and urinary L-FABP levels significantly decreased. CONCLUSION Histone is one of the causative agents for the increase of urinary L-FABP at an early stage of AKI. In addition, it suggested that urinary L-FABP may be useful as a subclinical AKI marker reflecting kidney damage induced by histone. Furthermore, urinary L-FABP reflected the degree of the damage after the administration of therapeutic agents such as heparin and PDE4 inhibitor.
Collapse
Affiliation(s)
- Keiichi Ohata
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- CMIC Holdings Co., Ltd, Tokyo, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | - Hanh Nhung Nguyen
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Karin Arai
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuri Hatanaka
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Kinuko Uno
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Marika Tohma
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Teppei Uechi
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Keita Sekiguchi
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Tsuyoshi Oikawa
- CMIC Holdings Co., Ltd, Tokyo, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | | | | | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Noriko Suzuki-Kemuriyama
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Dai Nakae
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
- Department of Medical Sports, Faculty of Health Care and Medical Sports, Teikyo Heisei University, Chiba, Japan
| | - Eisei Noiri
- National Center Biobank Network, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsuhiro Miyajima
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
14
|
Matsushita K, Toyoda T, Akane H, Morikawa T, Ogawa K. Role of CD44 expressed in renal tubules during maladaptive repair in renal fibrogenesis in an allopurinol-induced rat model of chronic kidney disease. J Appl Toxicol 2024; 44:455-469. [PMID: 37876353 DOI: 10.1002/jat.4554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/30/2023] [Accepted: 10/01/2023] [Indexed: 10/26/2023]
Abstract
The kidney is a major target organ for the adverse effects of pharmaceuticals; renal tubular epithelial cells (TECs) are particularly vulnerable to drug-induced toxicity. TECs have regenerative capacity; however, maladaptive repair of TECs after injury leads to renal fibrosis, resulting in chronic kidney disease (CKD). We previously reported the specific expression of CD44 in failed-repair TECs of rat CKD model induced by ischemia reperfusion injury. Here, we investigated the pathophysiological role of CD44 in renal fibrogenesis in allopurinol-treated rat CKD model. Dilated or atrophic TECs expressing CD44 in fibrotic areas were collected by laser microdissection and subjected to microarray analysis. Gene ontology showed that extracellular matrix (ECM)-related genes were upregulated and differentiation-related genes were downregulated in dilated/atrophic TECs. Ingenuity Pathway Analysis identified CD44 as an upstream regulator of fibrosis-related genes, including Fn1, which encodes fibronectin. Immunohistochemistry demonstrated that dilated/atrophic TECs expressing CD44 showed decreases in differentiation markers of TECs and clear expression of mesenchymal markers during basement membrane attachment. In situ hybridization revealed an increase in Fn1 mRNA in the cytoplasm of dilated/atrophic TECs, whereas fibronectin was localized in the stroma around these TECs, supporting the production/secretion of ECM by dilated/atrophic TECs. Overall, these data indicated that dilated/atrophic TECs underwent a partial epithelial-mesenchymal transition (pEMT) and that CD44 promoted renal fibrogenesis via induction of ECM production in failed-repair TECs exhibiting pEMT. CD44 was detected in the urine and serum of APL-treated rats, which may reflect the expression of CD44 in the kidney.
Collapse
Affiliation(s)
- Kohei Matsushita
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Hirotoshi Akane
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Tomomi Morikawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
15
|
DiKun KM, Tang XH, Fu L, Choi ME, Lu C, Gudas LJ. Retinoic acid receptor α activity in proximal tubules prevents kidney injury and fibrosis. Proc Natl Acad Sci U S A 2024; 121:e2311803121. [PMID: 38330015 PMCID: PMC10873609 DOI: 10.1073/pnas.2311803121] [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: 07/11/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024] Open
Abstract
Chronic kidney disease (CKD) is characterized by a gradual loss of kidney function and affects ~13.4% of the global population. Progressive tubulointerstitial fibrosis, driven in part by proximal tubule (PT) damage, is a hallmark of late stages of CKD and contributes to the development of kidney failure, for which there are limited treatment options. Normal kidney development requires signaling by vitamin A (retinol), which is metabolized to retinoic acid (RA), an endogenous agonist for the RA receptors (RARα, β, γ). RARα levels are decreased in a mouse model of diabetic nephropathy and restored with RA administration; additionally, RA treatment reduced fibrosis. We developed a mouse model in which a spatiotemporal (tamoxifen-inducible) deletion of RARα in kidney PT cells of adult mice causes mitochondrial dysfunction, massive PT injury, and apoptosis without the use of additional nephrotoxic substances. Long-term effects (3 to 4.5 mo) of RARα deletion include increased PT secretion of transforming growth factor β1, inflammation, interstitial fibrosis, and decreased kidney function, all of which are major features of human CKD. Therefore, RARα's actions in PTs are crucial for PT homeostasis, and loss of RARα causes injury and a key CKD phenotype.
Collapse
Affiliation(s)
- Krysta M. DiKun
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY10065
- Weill Cornell Graduate School of Medical Sciences, New York, NY10065
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY10065
| | - Leiping Fu
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY10065
| | - Mary E. Choi
- New York Presbyterian Hospital, New York, NY10065
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY10065
| | | | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY10065
- Weill Cornell Graduate School of Medical Sciences, New York, NY10065
- Department of Urology, New York, NY10065
| |
Collapse
|
16
|
Carlström M, Rannier Ribeiro Antonino Carvalho L, Guimaraes D, Boeder A, Schiffer TA. Dimethyl malonate preserves renal and mitochondrial functions following ischemia-reperfusion via inhibition of succinate dehydrogenase. Redox Biol 2024; 69:102984. [PMID: 38061207 PMCID: PMC10749277 DOI: 10.1016/j.redox.2023.102984] [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: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI), often experienced at the intensive care units, is associated with high morbidity/mortality where ischemia-reperfusion injury is a main causative factor. Succinate accumulation during ischemia contributes to the excessive generation of reactive oxygen species at reperfusion. Inhibition of succinate dehydrogenase has been associated with protective outcome in cardiac ischemia-reperfusion after 24h, but the effects on kidney and mitochondrial functions are less well studied. AIM To investigate the therapeutic potential of succinate dehydrogenase inhibition, by using dimethyl malonate (DMM), on kidney and mitochondria functions in a mouse model of AKI. METHODS Male C57BL/6J mice were pre-treated with DMM or placebo, i.p. 30min prior to bilateral renal ischemia (20min). After 3-days of reperfusion, glomerular filtration rate (GFR) was calculated from plasma clearance of FITC-inulin. Kidney mitochondria was isolated and mass specific and intrinsic mitochondrial function were evaluated by high resolution respirometry. Kidney sections were stained (i.e., hematoxylin-eosin and TUNEL) and analyzed for histopathological evaluation of injuries and apotosis, respectively. NADPH oxidase activity in kidney and human proximal tubular cell-line (HK2) were measured luminometrically. RESULTS DMM treatment improved GFR (p < 0.05) and reduced levels of blood urea nitrogen (p < 0.01) compared to untreated animals, which was associated with lower degree of ischemia-reperfusion-induced tubular injuries (P < 0.001) and apoptosis (P < 0.01). These therapeutic renal effects were linked with improved mitochondrial function, both mass-specific and intrinsic. Finally, DMM treatment prevented ischemia-reperfusion-induced NADPH oxidase activity in the kidney (p < 0.001), which was showed also in HK2 cells exposed to hypoxia and reoxygenation (P < 0.01). CONCLUSION Inhibition of succinate dehydrogenase with DMM, in conjunction with the ischemia-reperfusion phase, significantly improved both renal and mitochondrial functions. These findings may have clinical implications for future therapeutic strategies to prevent development of AKI and associated adverse complications, especially in high risk hospitalized patients.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Drielle Guimaraes
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ariela Boeder
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
17
|
Sonoda A, Shimada T, Saito K, Kosugi R, Taguchi Y, Inoue T. Light and Shadow of Na-Glucose Cotransporter 2 Inhibitors in the Treatment of Diabetes Mellitus: Points for Improvement Based on Our Clinical Experience. Int J Endocrinol 2024; 2024:3937927. [PMID: 38304078 PMCID: PMC10834091 DOI: 10.1155/2024/3937927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
We analyzed the effect of Na-glucose cotransporter 2 inhibitors (SGLT2-I) in diabetic patients visiting our hospital. The study included 236 patients treated with SGLT2-I alone or with codiabetic drugs for at least two years. We analyzed overtime changes in glycosylated hemoglobin A1c (HbA1c) in the patients by repeated analyses of variance (ANOVA) and evaluated the therapeutic effect. HbA1c levels decreased significantly in the first six months after treatment. Afterward, they leveled off and increased slightly over the next two years. Six months after treatment, the mean (SD) of HbA1c was 8.19 (1.46) %; the mean difference dropped by 0.91%, and HbA1c in mild DM2 did not drop by below 8.0%. Overall, there was only a slight improvement. We performed multivariate logistic regression analysis using a model with or without improvement as the objective variable and several explanatory variables. Na and Hct were significant factors. They increased considerably over six months and then leveled off. eGFR significantly reduced in the hyperfiltration group six months after treatment. The annual decline rate in eGFR was also faster, even in the nonhyperfiltration group than in the healthy subjects, which may be a characteristic of renal clearance in SGLT2-I treatment. In conclusion, SGLT2-I is an excellent antidiabetic, nephroprotective agent to eliminate hyperfiltration, but unfortunately, SGLT2-I alone does not have enough power to reduce blood glucose levels. SGLT2-I, with insulin or insulin secretagogues, enhances insulin resistance, induces hyperinsulinemia, and exacerbates type 2 DM. In contrast, SGLT2-I, with noninsulin antidiabetic agents and a low-carbohydrate diet, may bring better results.
Collapse
Affiliation(s)
- Akihiro Sonoda
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Toshio Shimada
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Clinical Laboratory, Shizuoka General Hospital, Shizuoka, Japan
| | - Kohei Saito
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Division of Diabetes, Endocrinology, and Metabolism, Shizuoka General Hospital, Shizuoka, Japan
| | - Rieko Kosugi
- Division of Diabetes, Endocrinology, and Metabolism, Shizuoka General Hospital, Shizuoka, Japan
| | - Yoshitaka Taguchi
- Division of Diabetes, Endocrinology, and Metabolism, Shizuoka General Hospital, Shizuoka, Japan
| | - Tatsuhide Inoue
- Division of Diabetes, Endocrinology, and Metabolism, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
18
|
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J, Liao L. Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne) 2024; 14:1256375. [PMID: 38260142 PMCID: PMC10801024 DOI: 10.3389/fendo.2023.1256375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
The energy needs of tubular epithelial components, especially proximal tubular epithelial cells (PTECs), are high and they heavily depend on aerobic metabolism. As a result, they are particularly vulnerable to various injuries caused by factors such as ischemia, proteinuria, toxins, and elevated glucose levels. Initial metabolic and phenotypic changes in PTECs after injury are likely an attempt at survival and repair. Nevertheless, in cases of recurrent or prolonged injury, PTECs have the potential to undergo a transition to a secretory state, leading to the generation and discharge of diverse bioactive substances, including transforming growth factor-β, Wnt ligands, hepatocyte growth factor, interleukin (IL)-1β, lactic acid, exosomes, and extracellular vesicles. By promoting fibroblast activation, macrophage recruitment, and endothelial cell loss, these bioactive compounds stimulate communication between epithelial cells and other interstitial cells, ultimately worsening renal damage. This review provides a summary of the latest findings on bioactive compounds that facilitate the communication between these cellular categories, ultimately leading to the advancement of tubulointerstitial fibrosis (TIF).
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yutian Tian
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| |
Collapse
|
19
|
Yamashita N, Kramann R. Mechanisms of kidney fibrosis and routes towards therapy. Trends Endocrinol Metab 2024; 35:31-48. [PMID: 37775469 DOI: 10.1016/j.tem.2023.09.001] [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: 07/18/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023]
Abstract
Kidney fibrosis is the final common pathway of virtually all chronic kidney diseases (CKDs) and is therefore considered to be a promising therapeutic target for these conditions. However, despite great progress in recent years, no targeted antifibrotic therapies for the kidney have been approved, likely because the complex mechanisms that initiate and drive fibrosis are not yet completely understood. Recent single-cell genomic approaches have allowed novel insights into kidney fibrosis mechanisms in mouse and human, particularly the heterogeneity and differentiation processes of myofibroblasts, the role of injured epithelial cells and immune cells, and their crosstalk mechanisms. In this review we summarize the key mechanisms that drive kidney fibrosis, including recent advances in understanding the mechanisms, as well as potential routes for developing novel targeted antifibrotic therapeutics.
Collapse
Affiliation(s)
- Noriyuki Yamashita
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology, and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
Zhi Y, Wu X, Chen Y, Chen X, Chen X, Luo H, Yi X, Lin X, Ma L, Chen Y, Cao Y, Li F, Zhou P. A novel TWIK2 channel inhibitor binds at the bottom of the selectivity filter and protects against LPS-induced experimental endotoxemia in vivo. Biochem Pharmacol 2023; 218:115894. [PMID: 37898389 DOI: 10.1016/j.bcp.2023.115894] [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: 08/26/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
TWIK2 channel plays a critical role in NLRP3 inflammasome activation and mice deficient in TWIK2 channel are protected from sepsis and inflammatory lung injury. However, inhibitors of TWIK2 channel are currently in an early stage of development, and the molecular determinants underlying the chemical modulation of TWIK2 channel remain unexplored. In this study, we identified NPBA and the synthesized derivative NPBA-4 potently and selectively inhibited TWIK2 channel by using whole-cell patch clamp techniques. Furthermore, the mutation of the last residues of the selectivity filter in both P1 and P2 (i.e., T106A, T214A) of TWIK2 channel substantially abolished the effect of NPBA on TWIK2 channel. Our data suggest that NPBA blocked TWIK2 channel through binding at the bottom of the selectivity filter, which was also supported by molecular docking prediction. Moreover, we found that NPBA significantly suppressed NLRP3 inflammasome activation in macrophages and alleviated LPS-induced endotoxemia and organ injury in vivo. Notably, the protective effects of NPBA against LPS-induced endotoxemia were abolished in Kcnk6-/- mice. In summary, our study has uncovered a series of novel inhibitors of TWIK2 channel and revealed their distinct molecular determinants interacting TWIK2 channel. These findings provide new insights into the mechanisms of pharmacological action on TWIK2 channel and opportunities for the development of selective TWIK2 channel modulators to treat related inflammatory diseases.
Collapse
Affiliation(s)
- Yuanxing Zhi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Xiaoyan Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanshan Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xingyuan Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiangyu Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hui Luo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Yi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiuling Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liang Ma
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yao Chen
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
21
|
Wang Y, Ran L, Lan Q, Liao W, Wang L, Wang Y, Xiong J, Li F, Yu W, Li Y, Huang Y, He T, Wang J, Zhao J, Yang K. Imbalanced lipid homeostasis caused by membrane αKlotho deficiency contributes to the acute kidney injury to chronic kidney disease transition. Kidney Int 2023; 104:956-974. [PMID: 37673285 DOI: 10.1016/j.kint.2023.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/28/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023]
Abstract
After acute kidney injury (AKI), renal tubular epithelial cells (RTECs) are pathologically characterized by intracellular lipid droplet (LD) accumulation, which are involved in RTEC injury and kidney fibrosis. However, its pathogenesis remains incompletely understood. The protein, αKlotho, primarily expressed in RTECs, is well known as an anti-aging hormone wielding versatile functions, and its membrane form predominantly acts as a co-receptor for fibroblast growth factor 23. Here, we discovered a connection between membrane αKlotho and intracellular LDs in RTECs. Fluorescent fatty acid (FA) pulse-chase assays showed that membrane αKlotho deficiency in RTECs, as seen in αKlotho homozygous mutated (kl/kl) mice or in mice with ischemia-reperfusion injury (IRI)-induced AKI, inhibited FA mobilization from LDs by impairing adipose triglyceride lipase (ATGL)-mediated lipolysis and lipophagy. This resulted in LD accumulation and FA underutilization. IRI-induced alterations were more striking in αKlotho deficiency. Mechanistically, membrane αKlotho deficiency promoted E3 ligase peroxin2 binding to ubiquitin-conjugating enzyme E2 D2, resulting in ubiquitin-mediated degradation of ATGL which is a common molecular basis for lipolysis and lipophagy. Overexpression of αKlotho rescued FA mobilization by preventing ATGL ubiquitination, thereby lessening LD accumulation and fibrosis after AKI. This suggests that membrane αKlotho is indispensable for the maintenance of lipid homeostasis in RTECs. Thus, our study identified αKlotho as a critical regulator of lipid turnover and homeostasis in AKI, providing a viable strategy for preventing tubular injury and the AKI-to-chronic kidney disease transition.
Collapse
Affiliation(s)
- Yue Wang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Ran
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qigang Lan
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Weinian Liao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yaqin Wang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiachuan Xiong
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fugang Li
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenrui Yu
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Li
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yinghui Huang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ting He
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Ke Yang
- Department of Nephrology, Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
22
|
Heyman SN, Armaly Z, Hamo-Giladi DB, Abassi Z. Novel perspectives regarding the physiologic mechanisms by which gliflozins induce reticulocytosis and erythrocytosis. Am J Physiol Endocrinol Metab 2023; 325:E621-E623. [PMID: 37819195 DOI: 10.1152/ajpendo.00277.2023] [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: 08/29/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023]
Abstract
Gliflozins provide a breakthrough in the management of type-2 diabetes. In addition to facilitating normoglycemia, these sodium-glucose cotransporter type 2 (SGLT2) inhibitors attenuate obesity, hypertension, dyslipidemia, and fluid retention, reduce cardiovascular morbidity, retard the progression of renal dysfunction, and improve survival. The administration of gliflozins also triggers erythropoietin (EPO) production, with the consequent induction of reticulocytosis and erythrocytosis. The mechanism(s) by which gliflozins induce erythropoiesis is a matter of debate. Whereas the canonical pathway of triggering EPO synthesis is through renal tissue hypoxia, it has been suggested that improved renal oxygenation may facilitate EPO synthesis via noncanonical trails. The latter proposes that recovery of peritubular interstitial fibroblasts producing erythropoietin (EPO) is responsible for enhanced erythropoiesis. According to this hypothesis, enhanced glucose/sodium reuptake by proximal tubules in uncontrolled diabetes generates cortical hypoxia, with injury to these cells. Once transport workload declines with the use of SGLT2i, they recover and regain their capacity to produce EPO. In this short communication, we argue that this hypothesis is incorrect. First, there is no evidence for interstitial cell injury related to hypoxia in the diabetic kidney. Tubular, rather than interstitial cells are prone to hypoxic injury in the diabetic kidney. Moreover, hypoxia, not normoxia, stimulates EPO synthesis by hypoxia-inducible factors (HIFs). Hypoxia regulates EPO synthesis as it blocks HIF prolyl hydroxylases (that initiate HIF alpha degradation), hence stabilizing HIF signals, inducing HIF-dependent genes, including EPO located in the deep cortex, and its production is initiated by the apocrinic formation of HIF-2, colocalized in these same cells.
Collapse
Affiliation(s)
- Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus and Herzog Hospital, Jerusalem, Israel
| | - Zaher Armaly
- Department of Nephrology, Nazareth Hospital-EMMS, Nazareth and the Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | - Zaid Abassi
- Ruth & Bruce Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel
- Department of Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
23
|
Zhong D, Quan L, Hao C, Chen J, Qiao R, Lin T, Ying C, Sun D, Jia Z, Sun Y. Targeting mPGES-2 to protect against acute kidney injury via inhibition of ferroptosis dependent on p53. Cell Death Dis 2023; 14:710. [PMID: 37907523 PMCID: PMC10618563 DOI: 10.1038/s41419-023-06236-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Acute kidney injury (AKI) is a clinical syndrome with high morbidity and mortality but no specific therapy. Microsomal prostaglandin E synthase-2 (mPGES-2) is a PGE2 synthase but can metabolize PGH2 to malondialdehyde by forming a complex with heme. However, the role and mechanism of action of mPGES-2 in AKI remain unclear. To examine the role of mPGES-2, both global and tubule-specific mPGES-2-deficient mice were treated with cisplatin to induce AKI. mPGES-2 knockdown or overexpressing HK-2 cells were exposed to cisplatin to cause acute renal tubular cell injury. The mPGES-2 inhibitor SZ0232 was used to test the translational potential of targeting mPGES-2 in treating AKI. Additionally, mice were subjected to unilateral renal ischemia/reperfusion to further validate the effect of mPGES-2 on AKI. Interestingly, both genetic and pharmacological blockage of mPGES-2 led to decreased renal dysfunction and morphological damage induced by cisplatin and unilateral renal ischemia/reperfusion. Mechanistic exploration indicated that mPGES-2 deficiency inhibited ferroptosis via the heme-dependent regulation of the p53/SLC7A11/GPX4 axis. The present study indicates that mPGES-2 blockage may be a promising therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Dandan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Lingling Quan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Chang Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Jingshuo Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, P. R. China
| | - Ranran Qiao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Tengfei Lin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Changjiang Ying
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221000, P. R. China
- Institute of Nephrology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
| | - Dong Sun
- Institute of Nephrology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, P. R. China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210008, P. R. China.
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, P. R. China.
| |
Collapse
|
24
|
Sung MJ, An HJ, Ha MH, Park SH, Jeong HY, Baek J, Lee SH, Lee YH, Lee SY. PTEN-induced kinase 1 exerts protective effects in diabetic kidney disease by attenuating mitochondrial dysfunction and necroptosis. Int J Biol Sci 2023; 19:5145-5159. [PMID: 37928264 PMCID: PMC10620829 DOI: 10.7150/ijbs.83906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in diabetic kidney disease initiation and progression. PTEN-induced serine/threonine kinase 1 (PINK1) is a core organizer of mitochondrial quality control; however, its function in diabetic kidney disease remains controversial. Here, we aimed to investigate the pathophysiological roles of PINK1 in diabetic tubulopathy, focusing on its effects on mitochondrial homeostasis and tubular cell necroptosis, which is a specialized form of regulated cell death. PINK1-knockout mice showed more severe diabetes-induced tubular injury, interstitial fibrosis, and albuminuria. The expression of profibrotic cytokines significantly increased in the kidneys of diabetic Pink1-/- mice, which eventually culminated in aggravated interstitial fibrosis. Additionally, the knockdown of PINK1 in HKC-8 cells upregulated the fibrosis-associated proteins, and these effects were rescued by PINK1 overexpression. PINK1 deficiency was also associated with exaggerated hyperglycemia-induced mitochondrial dysfunction and defective mitophagic activity, whereas PINK1 overexpression ameliorated these negative effects and restored mitochondrial homeostasis. Mitochondrial reactive oxygen species triggered tubular cell necroptosis under hyperglycemic conditions, which was aggravated by PINK1 deficiency and improved by its overexpression. In conclusion, PINK1 plays a pivotal role in suppressing mitochondrial dysfunction and tubular cell necroptosis under high glucose conditions and exerts protective effects in diabetic kidney disease.
Collapse
Affiliation(s)
- Min-Ji Sung
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Hyun-Ju An
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Min Heui Ha
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Seon Hwa Park
- Devision of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Hye Yun Jeong
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jihyun Baek
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Sang Ho Lee
- Devision of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Yu Ho Lee
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - So-Young Lee
- Devision of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| |
Collapse
|
25
|
Yoshimura Y, Muto Y, Omachi K, Miner JH, Humphreys BD. Elucidating the Proximal Tubule HNF4A Gene Regulatory Network in Human Kidney Organoids. J Am Soc Nephrol 2023; 34:1672-1686. [PMID: 37488681 PMCID: PMC10561821 DOI: 10.1681/asn.0000000000000197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/08/2023] [Indexed: 07/26/2023] Open
Abstract
SIGNIFICANCE STATEMENT HNF4 genes promote proximal tubule differentiation in mice, but their function in human nephrogenesis is not fully defined. This study uses human pluripotent stem cell (PSC)-derived kidney organoids as a model to investigate HNF4A and HNF4G functions. The loss of HNF4A , but not HNF4G , impaired reabsorption-related molecule expression and microvilli formation in human proximal tubules. Cleavage under targets and release using nuclease (CUT&RUN) sequencing and CRISPR-mediated transcriptional activation (CRISPRa) further confirm that HNF4A directly regulates its target genes. Human kidney organoids provide a good model for studying transcriptional regulation in human kidney development. BACKGROUND The proximal tubule plays a major role in electrolyte homeostasis. Previous studies have shown that HNF4A regulates reabsorption-related genes and promotes proximal tubule differentiation during murine kidney development. However, the functions and gene regulatory mechanisms of HNF4 family genes in human nephrogenesis have not yet been investigated. METHODS We generated HNF4A -knock out (KO), HNF4G -KO, and HNF4A/4G -double KO human pluripotent stem cell lines, differentiated each into kidney organoids, and used immunofluorescence analysis, electron microscopy, and RNA-seq to analyze them. We probed HNF4A-binding sites genome-wide by cleavage under targets and release using nuclease sequencing in both human adult kidneys and kidney organoid-derived proximal tubular cells. Clustered Regularly Interspaced Short Palindromic Repeats-mediated transcriptional activation validated HNF4A and HNF4G function in proximal tubules during kidney organoid differentiation. RESULTS Organoids lacking HNF4A , but not HNF4G , showed reduced expression of transport-related, endocytosis-related, and brush border-related genes, as well as disorganized brush border structure in the apical lumen of the organoid proximal tubule. Cleavage under targets and release using nuclease revealed that HNF4A primarily bound promoters and enhancers of genes that were downregulated in HNF4A -KO, suggesting direct regulation. Induced expression of HNF4A or HNF4G by CRISPR-mediated transcriptional activation drove increased expression of selected target genes during kidney organoid differentiation. CONCLUSIONS This study reveals regulatory mechanisms of HNF4A and HNF4G during human proximal tubule differentiation. The experimental strategy can be applied more broadly to investigate transcriptional regulation in human kidney development.
Collapse
Affiliation(s)
- Yasuhiro Yoshimura
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Kohei Omachi
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jeffrey H. Miner
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
26
|
Naas S, Schiffer M, Schödel J. Hypoxia and renal fibrosis. Am J Physiol Cell Physiol 2023; 325:C999-C1016. [PMID: 37661918 DOI: 10.1152/ajpcell.00201.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Renal fibrosis is the final stage of most progressive kidney diseases. Chronic kidney disease (CKD) is associated with high comorbidity and mortality. Thus, preventing fibrosis and thereby preserving kidney function increases the quality of life and prolongs the survival of patients with CKD. Many processes such as inflammation or metabolic stress modulate the progression of kidney fibrosis. Hypoxia has also been implicated in the pathogenesis of renal fibrosis, and oxygen sensing in the kidney is of outstanding importance for the body. The dysregulation of oxygen sensing in the diseased kidney is best exemplified by the loss of stimulation of erythropoietin production from interstitial cells in the fibrotic kidney despite anemia. Furthermore, hypoxia is present in acute or chronic kidney diseases and may affect all cell types present in the kidney including tubular and glomerular cells as well as resident immune cells. Pro- and antifibrotic effects of the transcription factors hypoxia-inducible factors 1 and 2 have been described in a plethora of animal models of acute and chronic kidney diseases, but recent advances in sequencing technologies now allow for novel and deeper insights into the role of hypoxia and its cell type-specific effects on the progression of renal fibrosis, especially in humans. Here, we review existing literature on how hypoxia impacts the development and progression of renal fibrosis.
Collapse
Affiliation(s)
- Stephanie Naas
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
27
|
Yang S, Yang G, Wang X, Xiang J, Kang L, Liang Z. SIRT2 alleviated renal fibrosis by deacetylating SMAD2 and SMAD3 in renal tubular epithelial cells. Cell Death Dis 2023; 14:646. [PMID: 37777567 PMCID: PMC10542381 DOI: 10.1038/s41419-023-06169-1] [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: 05/14/2023] [Revised: 09/06/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Transforming growth factor-β (TGF-β) is the primary factor that drives fibrosis in most, if not all, forms of chronic kidney disease. In kidneys that are obstructed, specific deletion of Sirt2 in renal tubule epithelial cells (TEC) has been shown to aggravate renal fibrosis, while renal tubule specific overexpression of Sirt2 has been shown to ameliorate renal fibrosis. Similarly, specific deletion of Sirt2 in hepatocyte aggravated CCl4-induced hepatic fibrosis. In addition, we have demonstrated that SIRT2 overexpression and knockdown restrain and enhance TGF-β-induced fibrotic gene expression, respectively, in TEC. Mechanistically, SIRT2 reduced the phosphorylation, acetylation, and nuclear localization levels of SMAD2 and SMAD3, leading to inhibition of the TGF-β signaling pathway. Further studies have revealed that that SIRT2 was able to directly interact with and deacetylate SMAD2 at lysine 451, promoting its ubiquitination and degradation. Notably, loss of SMAD specific E3 ubiquitin protein ligase 2 abolishes the ubiquitination and degradation of SMAD2 induced by SIRT2 in SMAD2. Regarding SMAD3, we have found that SIRT2 interact with and deacetylates SMAD3 at lysine 341 and 378 only in the presence of TGF-β, thereby reducing its activation. This study provides initial indication of the anti-fibrotic role of SIRT2 in renal tubules and hepatocytes, suggesting its therapeutic potential for fibrosis.
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Guangyan Yang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Xinyu Wang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Jiaqing Xiang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China
| | - Lin Kang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People's Hospital, Southern University of Science and Technology, Shenzhen, China.
| | - Zhen Liang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University & The First Affiliated Hospital of Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
28
|
Dai ZC, Chen JX, Zou R, Liang XB, Tang JX, Yao CW. Role and mechanisms of SGLT-2 inhibitors in the treatment of diabetic kidney disease. Front Immunol 2023; 14:1213473. [PMID: 37809091 PMCID: PMC10552262 DOI: 10.3389/fimmu.2023.1213473] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Diabetic kidney disease (DKD) is a chronic inflammatory condition that affects approximately 20-40% of individuals with diabetes. Sodium-glucose co-transporter 2 (SGLT-2) inhibitors, emerging as novel hypoglycemic agents, have demonstrated significant cardiorenal protective effects in patients with DKD. Initially, it was believed that the efficacy of SGLT-2 inhibitors declined as the estimated glomerular filtration rate (eGFR) decreased, which led to their preferential use in DKD patients at G1-G3 stages. However, recent findings from the DAPA-CKD and EMPA-KIDNEY studies have revealed equally beneficial cardiorenal effects of SGLT-2 inhibitors in individuals at stage G4 DKD, although the underlying mechanism behind this phenomenon remains unclear. In this comprehensive analysis, we provide a systematic review of the mechanisms and functioning of SGLT-2 inhibitors, potential renal protection mechanisms, and the therapeutic efficacy and safety of SGLT-2 inhibitors in kidney diseases, with a particular focus on stage G4 DKD. Gaining a deeper understanding of the renal protective effect of SGLT-2 inhibitors and their underlying mechanisms is highly significance for the successful utilization of these inhibitors in the treatment of diverse kidney disorders.
Collapse
Affiliation(s)
| | | | | | | | - Ji-Xin Tang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cui-Wei Yao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Diseases of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
29
|
Niculae A, Gherghina ME, Peride I, Tiglis M, Nechita AM, Checherita IA. Pathway from Acute Kidney Injury to Chronic Kidney Disease: Molecules Involved in Renal Fibrosis. Int J Mol Sci 2023; 24:14019. [PMID: 37762322 PMCID: PMC10531003 DOI: 10.3390/ijms241814019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is one of the main conditions responsible for chronic kidney disease (CKD), including end-stage renal disease (ESRD) as a long-term complication. Besides short-term complications, such as electrolyte and acid-base disorders, fluid overload, bleeding complications or immune dysfunctions, AKI can develop chronic injuries and subsequent CKD through renal fibrosis pathways. Kidney fibrosis is a pathological process defined by excessive extracellular matrix (ECM) deposition, evidenced in chronic kidney injuries with maladaptive architecture restoration. So far, cited maladaptive kidney processes responsible for AKI to CKD transition were epithelial, endothelial, pericyte, macrophage and fibroblast transition to myofibroblasts. These are responsible for smooth muscle actin (SMA) synthesis and abnormal renal architecture. Recently, AKI progress to CKD or ESRD gained a lot of interest, with impressive progression in discovering the mechanisms involved in renal fibrosis, including cellular and molecular pathways. Risk factors mentioned in AKI progression to CKD are frequency and severity of kidney injury, chronic diseases such as uncontrolled hypertension, diabetes mellitus, obesity and unmodifiable risk factors (i.e., genetics, older age or gender). To provide a better understanding of AKI transition to CKD, we have selected relevant and updated information regarding the risk factors responsible for AKIs unfavorable long-term evolution and mechanisms incriminated in the progression to a chronic state, along with possible therapeutic approaches in preventing or delaying CKD from AKI.
Collapse
Affiliation(s)
- Andrei Niculae
- Department of Nephrology, Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihai-Emil Gherghina
- Department of Nephrology, Ilfov County Emergency Clinical Hospital, 022104 Bucharest, Romania
| | - Ileana Peride
- Department of Nephrology, Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mirela Tiglis
- Department of Anesthesia and Intensive Care, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| | - Ana-Maria Nechita
- Department of Nephrology, “St. John” Emergency Clinical Hospital, 042122 Bucharest, Romania
| | | |
Collapse
|
30
|
Oh H, You JS, Bae H, Park GB, Chung YE. Delivery of recombinant sestrin2 ameliorates oxidative stress, mitochondrial damage and renal dysfunction in contrast-induced acute kidney injury. Biochem Pharmacol 2023; 215:115761. [PMID: 37604294 DOI: 10.1016/j.bcp.2023.115761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023]
Abstract
Although the use of iodinated contrast agents is sometimes unavoidable for accurate diagnosis, contrast-induced acute kidney injury (CI-AKI) is a possible complication of its administration. The pathogenesis of CI-AKI has not been fully elucidated, but oxidative stress is thought to be a major factor. Sestrin2 plays an important role in cellular and mitochondrial homeostasis by regulating oxidative stress. In this study, we aimed to investigate whether recombinant adenovirus containing sestrin2 (RS) can attenuate CI-AKI by reducing oxidative stress in a CI-AKI mice model. Our results showed that RS decreases oxidative stress, pro-inflammatory cytokines (TNF-α, IL-1α, IL-1β and IL-6) and apoptosis (Bax/Bcl2 and cleaved caspase-3) in the CI-AKI model. Additionally, RS alleviated mitochondrial damage, as evidenced by morphological changes, are restored ATP synthesis. Furthermore, RS administration resulted in a decrease in mitochondrial fission marker (Drp1) that was increased in the CI-AKI model, while the mitochondrial fusion marker (Mfn2) increased, indicating a restoration of mitochondrial dynamics. Decreased relative blood volume, as evaluated on computed tomography (CT), significantly increased compared to the CI-AKI group after RS administration. Finally, renal injury markers such as Kim-1, Ngal, IL-18 also decreased and kidney function was preserved with RS. These results suggested that RS can mitigate the deterioration of renal function in CI-AKI model.
Collapse
Affiliation(s)
- Hyewon Oh
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heejin Bae
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ga Bin Park
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
31
|
DiKun KM, Gudas LJ. Vitamin A and retinoid signaling in the kidneys. Pharmacol Ther 2023; 248:108481. [PMID: 37331524 PMCID: PMC10528136 DOI: 10.1016/j.pharmthera.2023.108481] [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: 04/03/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Vitamin A (VA, retinol) and its metabolites (commonly called retinoids) are required for the proper development of the kidney during embryogenesis, but retinoids also play key roles in the function and repair of the kidney in adults. Kidneys filter 180-200 liters of blood per day and each kidney contains approximately 1 million nephrons, which are often referred to as the 'functional units' of the kidney. Each nephron consists of a glomerulus and a series of tubules (proximal tubule, loop of Henle, distal tubule, and collecting duct) surrounded by a network of capillaries. VA is stored in the liver and converted to active metabolites, most notably retinoic acid (RA), which acts as an agonist for the retinoic acid receptors ((RARs α, β, and γ) to regulate gene transcription. In this review we discuss some of the actions of retinoids in the kidney after injury. For example, in an ischemia-reperfusion model in mice, injury-associated loss of proximal tubule (PT) differentiation markers occurs, followed by re-expression of these differentiation markers during PT repair. Notably, healthy proximal tubules express ALDH1a2, the enzyme that metabolizes retinaldehyde to RA, but transiently lose ALDH1a2 expression after injury, while nearby myofibroblasts transiently acquire RA-producing capabilities after injury. These results indicate that RA is important for renal tubular injury repair and that compensatory mechanisms exist for the generation of endogenous RA by other cell types upon proximal tubule injury. ALDH1a2 levels also increase in podocytes, epithelial cells of the glomeruli, after injury, and RA promotes podocyte differentiation. We also review the ability of exogenous, pharmacological doses of RA and receptor selective retinoids to treat numerous kidney diseases, including kidney cancer and diabetic kidney disease, and the emerging genetic evidence for the importance of retinoids and their receptors in maintaining or restoring kidney function after injury. In general, RA has a protective effect on the kidney after various types of injuries (eg. ischemia, cytotoxic actions of chemicals, hyperglycemia related to diabetes). As more research into the actions of each of the three RARs in the kidney is carried out, a greater understanding of the actions of vitamin A is likely to lead to new insights into the pathology of kidney disorders and the development of new therapies for kidney diseases.
Collapse
Affiliation(s)
- Krysta M DiKun
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Urology, Weill Cornell Medicine, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
32
|
Li Y, Gu W, Hepokoski M, Pham H, Tham R, Kim YC, Simonson TS, Singh P. Energy Metabolism Dysregulation in Chronic Kidney Disease. KIDNEY360 2023; 4:1080-1094. [PMID: 37222594 PMCID: PMC10476685 DOI: 10.34067/kid.0000000000000153] [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: 09/01/2022] [Accepted: 05/08/2023] [Indexed: 05/25/2023]
Abstract
Key Points There is significant enrichment in metabolic pathways in early stages in the subtotal nephrectomy model of CKD. Proximal tubular mitochondrial respiration is suppressed likely from mitochondrial dysfunction in substrate utilization and ATP synthesis. There is significant suppression of pyruvate dehydrogenase and increased glycolysis in proximal tubules. Background CKD is a significant contributor to morbidity and mortality. A better understanding of mechanisms underlying CKD progression is indispensable for developing effective therapies. Toward this goal, we addressed specific gaps in knowledge regarding tubular metabolism in the pathogenesis of CKD using the subtotal nephrectomy (STN) model in mice. Methods Weight- and age‐matched male 129X1/SvJ mice underwent sham or STN surgeries. We conducted serial GFR and hemodynamic measurements up to 16 weeks after sham and STN surgery and established the 4-week time point for subsequent studies. Results For a comprehensive assessment of renal metabolism, we conducted transcriptomic analyses, which showed significant enrichment of pathways involved in fatty acid metabolism, gluconeogenesis, glycolysis, and mitochondrial metabolism in STN kidneys. Expression of rate-limiting fatty acid oxidation and glycolytic enzymes was increased in STN kidneys, and proximal tubules in STN kidneys exhibited increased functional glycolysis but decreased mitochondrial respiration, despite an increase in mitochondrial biogenesis. Assessment of the pyruvate dehydrogenase complex pathway showed significant suppression of pyruvate dehydrogenase, suggesting decreased provision of acetyl CoA from pyruvate for the citric acid cycle to fuel mitochondrial respiration. Conclusion Metabolic pathways are significantly altered in response to kidney injury and may play an important role in the disease progression.
Collapse
Affiliation(s)
- Ying Li
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, California
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, California
| | - Hai Pham
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Rick Tham
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Young Chul Kim
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, San Diego, California
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California
- VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
33
|
Heyman SN, Abassi Z. Gliflozins, Erythropoietin, and Erythrocytosis: Is It Renal Normoxia- or Hypoxia-Driven? J Clin Med 2023; 12:4871. [PMID: 37510986 PMCID: PMC10381125 DOI: 10.3390/jcm12144871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The introduction of gliflozins in the management of type 2 diabetes mellitus leads to a better control of hyperglycemia, obesity, hypertension, dyslipidemia, and fluid retention. Most importantly, it also improves renal survival and reduces major cardiovascular events and mortality. Gliflozins were also found to induce erythropoietin (EPO) synthesis, leading to reticulocytosis and erythropoiesis. The mechanism(s) by which gliflozins induce erythropoiesis is a matter of debate. Although the canonical pathway of triggering EPO synthesis is through renal tissue hypoxia, it has been suggested that improved renal oxygenation may facilitate EPO synthesis via non-canonical routes. The latter proposes that the recovery of peritubular interstitial fibroblasts producing erythropoietin (EPO) is responsible for enhanced erythropoiesis. According to this hypothesis, enhanced glucose/sodium re-uptake by proximal tubules in uncontrolled diabetes generates cortical hypoxia, with injury to these cells. Once transport workload declines with the use of SGLT2i, they recover and regain their capacity to produce EPO. In this short communication, we argue that this hypothesis may be wrong and propose that gliflozins likely induce EPO through the documented intensification of renal hypoxia at the corticomedullary junction, related to the translocation of tubular transport from cortical segments to medullary thick ascending limbs. We propose that gliflozins, through intensified hypoxia in this region, trigger local EPO synthesis in peritubular interstitial cells via the canonical pathway of blocking HIF-prolyl hydroxylases (that initiate HIF alpha degradation), with the consequent stabilization of HIF-2 signal and an apocrinic induction of EPO in these same cells.
Collapse
Affiliation(s)
- Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus and Herzog Hospital, Jerusalem 9765422, Israel
| | - Zaid Abassi
- Department of Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
- Department of Physiology & Biophysics, The Rappaport Faculty of Medicine, Technion IIT, Haifa 3200003, Israel
| |
Collapse
|
34
|
Singh AK. Dapafliglozin and Correction of Anemia in Patients with CKD. NEJM EVIDENCE 2023; 2:EVIDe2300095. [PMID: 38320136 DOI: 10.1056/evide2300095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Anemia of chronic kidney disease (CKD) develops as kidney function declines. Reduced erythropoietin production, iron deficiency, and inflammation are the most important causes of CKD anemia. Anemia in the healthy population is defined by World Health Organization (WHO) criteria: for women, a hemoglobin (Hb) of less than 12 g/dl, and for men, an Hb of less than 13 g/dl. However, for practical purposes, severe anemia in patients with CKD requiring treatment is defined by a threshold Hb of less than 10 g/dl.
Collapse
Affiliation(s)
- Ajay K Singh
- Brigham and Women's Hospital, Harvard Medical School, Boston
| |
Collapse
|
35
|
Yang S, Xiang J, Ma C, Yang G, Wang X, Liu H, Fan G, Kang L, Liang Z. Sp1-like protein KLF13 acts as a negative feedback regulator of TGF-β signaling and fibrosis. Cell Rep 2023; 42:112367. [PMID: 37029927 DOI: 10.1016/j.celrep.2023.112367] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/23/2023] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
Transforming growth factor β (TGF-β) is the primary factor that drives fibrosis in most forms of chronic kidney disease. The aim of this study was to identify endogenous regulators of TGF-β signaling and fibrosis. Here, we show that tubulointerstitial fibrosis is aggravated by global deletion of KLF13 and attenuated by adeno-associated virus-mediated KLF13 overexpression in renal tubular epithelial cells. KLF13 recruits a repressor complex comprising SIN3A and histone deacetylase 1 (HDAC1) to the TGF-β target genes, limiting the profibrotic effects of TGF-β. Temporary upregulation of TGF-β induces KLF13 expression, creating a negative feedback loop that triggers the anti-fibrotic effect of KLF13. However, persistent activation of TGF-β signaling reduces KLF13 levels through FBXW7-mediated ubiquitination degradation and HDAC-dependent mechanisms to inhibit KLF13 transcription and offset the anti-fibrotic effect of KLF13. Collectively, our data demonstrate a role of KLF13 in regulating TGF-β signaling and fibrosis.
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Guangyan Yang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xinyu Wang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Hanyong Liu
- Department of Nephrology, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| | - Lin Kang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China; The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Zhen Liang
- Department of Geriatrics, The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| |
Collapse
|
36
|
Tanaka S, Wakui H, Azushima K, Tsukamoto S, Yamaji T, Urate S, Suzuki T, Abe E, Taguchi S, Yamada T, Kobayashi R, Kanaoka T, Kamimura D, Kinguchi S, Takiguchi M, Funakoshi K, Yamashita A, Ishigami T, Tamura K. Effects of a High-Protein Diet on Kidney Injury under Conditions of Non-CKD or CKD in Mice. Int J Mol Sci 2023; 24:ijms24097778. [PMID: 37175483 PMCID: PMC10177820 DOI: 10.3390/ijms24097778] [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: 03/02/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Considering the prevalence of obesity and global aging, the consumption of a high-protein diet (HPD) may be advantageous. However, an HPD aggravates kidney dysfunction in patients with chronic kidney disease (CKD). Moreover, the effects of an HPD on kidney function in healthy individuals are controversial. In this study, we employed a remnant kidney mouse model as a CKD model and aimed to evaluate the effects of an HPD on kidney injury under conditions of non-CKD and CKD. Mice were divided into four groups: a sham surgery (sham) + normal diet (ND) group, a sham + HPD group, a 5/6 nephrectomy (Nx) + ND group and a 5/6 Nx + HPD group. Blood pressure, kidney function and kidney tissue injury were compared after 12 weeks of diet loading among the four groups. The 5/6 Nx groups displayed blood pressure elevation, kidney function decline, glomerular injury and tubular injury compared with the sham groups. Furthermore, an HPD exacerbated glomerular injury only in the 5/6 Nx group; however, an HPD did not cause kidney injury in the sham group. Clinical application of these results suggests that patients with CKD should follow a protein-restricted diet to prevent the exacerbation of kidney injury, while healthy individuals can maintain an HPD without worrying about the adverse effects.
Collapse
Affiliation(s)
- Shohei Tanaka
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Takahiro Yamaji
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Shingo Urate
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Toru Suzuki
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Eriko Abe
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Shinya Taguchi
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Takayuki Yamada
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Ryu Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Daisuke Kamimura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Masahito Takiguchi
- Department of Neuroanatomy, School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kengo Funakoshi
- Department of Neuroanatomy, School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Akio Yamashita
- Department of Investigative Medicine, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishiharacho, Okinawa 903-0215, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
37
|
Chi X, Yang X, Li G, Wu H, Huang J, Qi Y, Tang G. A Comparative Study of 18F-FAPI-42 and 18F-FDG PET/CT for Evaluating Acute Kidney Injury in Cancer Patients. Mol Imaging Biol 2023:10.1007/s11307-023-01820-x. [PMID: 37020127 DOI: 10.1007/s11307-023-01820-x] [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: 01/31/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023]
Abstract
PURPOSE Compare the value of imaging using positron 18F-labeled fibroblast activation protein inhibitor-42 (18F-FAPI-42) and 18F-labeled deoxyglucose (18F-FDG) for assessment of AKI. PROCEDURES This study analyzed cancer patients who received 18F-FAPI-42 and 18F-FDG PET/CT imaging. Eight patients had AKI with bilateral ureteral obstruction (BUO), eight had BUO (CKD1-2) with no acute kidney disease (AKD), and eight had no ureteral obstruction (UO) with normal renal function. The average standardized uptake value (SUVave) of the renal parenchyma (RP-SUVave), the blood pool SUVave (B- SUVave), SUVave in the highest region of the renal collective system (RCS-SUVave), and the highest serum creatinine level (top SCr) were recorded. RESULTS The 18F-FAPI-42 and 18F-FDG results showed that radiotracer of renal parenchyma was more concentrated in the AKI group than in the other two groups, whereas the RP-SUVave from 18F-FAPI-42 was higher than that from 18F-FDG in the AKI group (all P < 0.05). 18F-FAPI-42 imaging in the AKI group showed uptake by the renal parenchyma with a diffuse increase, but very little radiotracer in the renal collecting system, similar to a "super kidney scan." The renal parenchyma also had an increase of SUVave, with accumulation of radiotracer in the renal collecting system. AKI was more severe when a patient had a "super kidney scan" in both kidneys (P < 0.05). The B-SUVave level was higher in the AKI group than in the other two groups in 18F-FAPI-42 (both P < 0.05). CONCLUSIONS 18F-FAPI-42 imaging had higher RP-SUVave than 18F-FDG imaging in cancer patients who had BUO with AKI. An increased renal parenchyma uptake in both kidneys and low radiotracer distribution in the collecting system suggest more severe AKI.
Collapse
Affiliation(s)
- Xiaohua Chi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiaoqiang Yang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Guiping Li
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Hubing Wu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jiawen Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Yongshuai Qi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Ganghua Tang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
38
|
Emre Aydıngöz S, Teimoori A, Orhan HG, Efe OE, Kibaroğlu S, Erdem ŞR. Effect of hydrogen sulfide on ischemia-reperfusion injury of kidney: A systematic review and meta-analysis of in vivo animal studies. Eur J Pharmacol 2023; 943:175564. [PMID: 36736943 DOI: 10.1016/j.ejphar.2023.175564] [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: 11/14/2022] [Revised: 01/14/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Hydrogen sulfide (H2S) has been shown to be effective against kidney ischemia-reperfusion injury (IRI) in animal studies. We aimed to evaluate the current evidence from in vivo animal studies for the protective effects of H2S against kidney IRI by systematically reviewing the literature and performing a meta-analysis. Based on the preregistered protocol (PROSPERO: CRD42021295469); PubMed, Medline, Embase, Web of Science, and Scopus were searched to identify in vivo animal studies evaluating the effect of H2S against kidney IRI. Standardized mean difference (SMD) with 95% confidence interval (CI) was calculated and pooled using random-effects meta-analysis. Twenty-two articles complied with eligibility criteria, from which the creatinine levels of 152 control animals and 182 animals treated with H2S from 27 individual experiments were pooled. H2S treatment significantly decreased serum creatinine (SMD = -1.82 [95% CI -1.12, -2.51], p < 0.0001), blood urea nitrogen (-2.50 [-1.46, -3.54], p < 0.0001), tissue malondialdehyde (-2.59 [-3.30, -1.88], p < 0.0001), tunel positive cells (-3.16 [-4.38, -1.94], p < 0.0001), and tubular damage score (-2.01 [-3.03, -0.99], p < 0.0001). There was a high heterogeneity across studies (I2 = 83.5% for serum creatinine level). In meta-regression analysis, the type of H2S donor and its application time accounted for 11.3% (p = 0.025) and 16.6% (p = 0.039) of heterogeneity, respectively. Accordingly, H2S protects the kidney against IRI only if it is given as GYY4137 before or during ischemia. Although H2S is a potential candidate against kidney IRI, further well-designed preclinical studies focusing on GYY4137 are warranted before clinical implication.
Collapse
Affiliation(s)
- Selda Emre Aydıngöz
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey.
| | - Arıyan Teimoori
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Halit Güner Orhan
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Oğuzhan Ekin Efe
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Seda Kibaroğlu
- Department of Pharmacology, Başkent University Institute of Health Sciences, Ankara, Turkey
| | - Ş Remzi Erdem
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
39
|
Reinhart GA, Harrison PC, Lincoln K, Chen H, Sun P, Hill J, Qian HS, McHugh MC, Clifford H, Ng KJ, Wang H, Fowler D, Gueneva-Boucheva K, Brenneman JB, Bosanac T, Wong D, Fryer RM, Sarko C, Boustany-Kari CM, Pullen SS. The Novel, Clinical-Stage Soluble Guanylate Cyclase Activator BI 685509 Protects from Disease Progression in Models of Renal Injury and Disease. J Pharmacol Exp Ther 2023; 384:382-392. [PMID: 36507845 DOI: 10.1124/jpet.122.001423] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/01/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Activation of soluble guanylate cyclase (sGC) to restore cyclic guanosine monophosphate (cGMP) and improve functionality of nitric oxide (NO) pathways impaired by oxidative stress is a potential treatment of diabetic and chronic kidney disease. We report the pharmacology of BI 685509, a novel, orally active small molecule sGC activator with disease-modifying potential. BI 685509 and human sGC α1/β1 heterodimer containing a reduced heme group produced concentration-dependent increases in cGMP that were elevated modestly by NO, whereas heme-free sGC and BI 685509 greatly enhanced cGMP with no effect of NO. BI 685509 increased cGMP in human and rat platelet-rich plasma treated with the heme-oxidant ODQ; respective EC50 values were 467 nM and 304 nM. In conscious telemetry-instrumented rats, BI 685509 did not affect mean arterial pressure (MAP) or heart rate (HR) at 3 and 10 mg/kg (p.o.), whereas 30 mg/kg decreased MAP and increased HR. Ten days of BI 685509 at supratherapeutic doses (60 or 100 mg/kg p.o., daily) attenuated MAP and HR responses to a single 100 mg/kg challenge. In the ZSF1 rat model, BI 685509 (1, 3, 10, and 30 mg/kg per day, daily) coadministered with enalapril (3 mg/kg per day) dose-dependently reduced proteinuria and incidence of glomerular sclerosis; MAP was modestly reduced at the higher doses versus enalapril. In the 7-day rat unilateral ureteral obstruction model, BI 685509 dose-dependently reduced tubulointerstitial fibrosis (P < 0.05 at 30 mg/kg). In conclusion, BI 685509 is a potent, orally bioavailable sGC activator with clear renal protection and antifibrotic activity in preclinical models of kidney injury and disease. SIGNIFICANCE STATEMENT: BI 685509 is a novel small soluble guanylate cyclase (sGC) molecule activator that exhibits an in vitro profile consistent with that of an sGC activator. BI 685509 reduced proteinuria and glomerulosclerosis in the ZSF1 rat, a model of diabetic kidney disease (DKD), and reduced tubulointerstitial fibrosis in a rat 7-day unilateral ureteral obstruction model. Thus, BI 685509 is a promising new therapeutic agent and is currently in phase II clinical trials for chronic kidney disease and DKD.
Collapse
Affiliation(s)
- Glenn A Reinhart
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Paul C Harrison
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Kathleen Lincoln
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Hongxing Chen
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Peng Sun
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Jon Hill
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Hu Sheng Qian
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Mark C McHugh
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Holly Clifford
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Khing Jow Ng
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Hong Wang
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Danielle Fowler
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Kristina Gueneva-Boucheva
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Jehrod B Brenneman
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Todd Bosanac
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Diane Wong
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Ryan M Fryer
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Chris Sarko
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Carine M Boustany-Kari
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| | - Steven S Pullen
- Departments of Cardiometabolic Diseases Research (G.A.R., P.C.H., K.L., H.C., P.S., H.S.Q., M.C.M., H.C., K.J.N., H.W., D.F., R.M.F., C.M.B.-K., S.S.P.), Small Molecule Discovery Research (K.G.-B., J.B.B., T.B., D.W., C.S.), and Global Computational Biology and Data Sciences (J.H.), Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut
| |
Collapse
|
40
|
Rodrigues CE, Endre ZH. Definitions, phenotypes, and subphenotypes in acute kidney injury-Moving towards precision medicine. Nephrology (Carlton) 2023; 28:83-96. [PMID: 36370326 PMCID: PMC10100386 DOI: 10.1111/nep.14132] [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: 06/24/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
The current definition of acute kidney injury (AKI) is generic and, based only on markers of function, is unsuitable for guiding individualized treatment. AKI is a complex syndrome with multiple presentations and causes. Targeted AKI management will only be possible if different phenotypes and subphenotypes of AKI are recognised, based on causation and related pathophysiology. Molecular signatures to identify subphenotypes are being recognised, as specific biomarkers reveal activated pathways. Assessment of individual clinical risk needs wider dissemination to allow identification of patients at high risk of AKI. New and more timely markers for glomerular filtration rate (GFR) are available. However, AKI diagnosis and classification should not be limited to GFR, but include tubular function and damage. Combining damage and stress biomarkers with functional markers enhances risk prediction, and identifies a population enriched for clinical trials targeting AKI. We review novel developments and aim to encourage implementation of these new techniques into clinical practice as a strategy for individualizing AKI treatment akin to a precision medicine-based approach.
Collapse
Affiliation(s)
- Camila Eleuterio Rodrigues
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
- Nephrology DepartmentHospital das Clínicas – University of São Paulo School of MedicineSão PauloBrazil
| | - Zoltán H. Endre
- Nephrology DepartmentPrince of Wales Clinical School – UNSW MedicineSydneyNew South WalesAustralia
| |
Collapse
|
41
|
Ekanayake P, Mudaliar S. Increase in hematocrit with SGLT-2 inhibitors - Hemoconcentration from diuresis or increased erythropoiesis after amelioration of hypoxia? Diabetes Metab Syndr 2023; 17:102702. [PMID: 36657305 DOI: 10.1016/j.dsx.2022.102702] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS The SGLT2-inhibitors significantly reduce heart failure hospitalization and progression to end-stage kidney disease. An increase in hemoglobin/hematocrit is seen with SGLT2i-inhibitor treatment. This increase has been attributed to hemoconcentration resulting from a diuretic effect. In this review, we present evidence suggesting that the hematocrit increase is not due to hemoconcentration, but to an increase in erythropoiesis due to amelioration of hypoxia and more efficient erythropoietin production with SGLT2-inhibitor treatment. METHODS We performed a detailed review of the literature in PubMed for articles describing various mechanisms linking hematocrit increase with SGLT2-inhibitor use to their cardio-renal benefits. RESULTS The best predictor of cardio-renal benefits with SGLT2-inhibitors is an increase in hematocrit and hemoglobin. If this hemoconcentration is a results of diuresis, this would be associated with volume contraction and a deterioration in renal function, as seen with long-term diuretic use. This is the opposite of what is seen with the use of SGLT2-inhibitors, which are associated with long-term preservation of renal function. There is now growing evidence that the increase in hematocrit can be attributed to an increase in erythropoiesis due to amelioration of renal hypoxia and more efficient erythropoietin production with SGLT2-inhibitor treatment. Increased erythropoiesis leads to an increase in RBC count which improves myocardial/renal tissue oxygenation and function. CONCLUSION The increase in hematocrit with SGLT2i treatment is not due to hemoconcentration, but to an increase in erythropoiesis due to amelioration of hypoxia and more efficient erythropoietin production with SGLT2i treatment.
Collapse
Affiliation(s)
- Preethika Ekanayake
- Veterans Affairs Medical Center, San Diego, CA, USA; Department of Medicine, University of California, San Diego School of Medicine, USA
| | - Sunder Mudaliar
- Veterans Affairs Medical Center, San Diego, CA, USA; Department of Medicine, University of California, San Diego School of Medicine, USA.
| |
Collapse
|
42
|
Peng Z, Wang H, Zheng J, Wang J, Xiang Y, Liu C, Ji M, Liu H, Pan L, Qin X, Qu X. Is the proximal tubule the focus of tubulointerstitial fibrosis? Heliyon 2023; 9:e13508. [PMID: 36846656 PMCID: PMC9950842 DOI: 10.1016/j.heliyon.2023.e13508] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Tubulointerstitial fibrosis (TIF), a common end result of almost all progressive chronic kidney diseases (CKD), is also the best predictor of kidney survival. Almost all cells in the kidney are involved in the progression of TIF. Myofibroblasts, the primary producers of extracellular matrix, have previously received a great deal of attention; however, a large body of emerging evidence reveals that proximal tubule (PT) plays a central role in TIF progression. In response to injury, renal tubular epithelial cells (TECs) transform into inflammatory and fibroblastic cells, producing various bioactive molecules that drive interstitial inflammation and fibrosis. Here we reviewed the increasing evidence for the key role of the PT in promoting TIF in tubulointerstitial and glomerular injury and discussed the therapeutic targets and carrier systems involving the PT that holds particular promise for treating patients with fibrotic nephropathy.
Collapse
Affiliation(s)
- Zhi Peng
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Hui Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Jiaoyun Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Chi Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Ming Ji
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Lang Pan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
43
|
Livingston MJ, Shu S, Fan Y, Li Z, Jiao Q, Yin XM, Venkatachalam MA, Dong Z. Tubular cells produce FGF2 via autophagy after acute kidney injury leading to fibroblast activation and renal fibrosis. Autophagy 2023; 19:256-277. [PMID: 35491858 PMCID: PMC9809951 DOI: 10.1080/15548627.2022.2072054] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Following acute kidney injury (AKI), renal tubular cells may stimulate fibroblasts in a paracrine fashion leading to interstitial fibrosis, but the paracrine factors and their regulation under this condition remain elusive. Here we identify a macroautophagy/autophagy-dependent FGF2 (fibroblast growth factor 2) production in tubular cells. Upon induction, FGF2 acts as a key paracrine factor to activate fibroblasts for renal fibrosis. After ischemic AKI in mice, autophagy activation persisted for weeks in renal tubular cells. In inducible, renal tubule-specific atg7 (autophagy related 7) knockout (iRT-atg7-KO) mice, autophagy deficiency induced after AKI suppressed the pro-fibrotic phenotype in tubular cells and reduced fibrosis. Among the major cytokines, tubular autophagy deficiency in iRT-atg7-KO mice specifically diminished FGF2. Autophagy inhibition also attenuated FGF2 expression in TGFB1/TGF-β1 (transforming growth factor, beta 1)-treated renal tubular cells. Consistent with a paracrine action, the culture medium of TGFB1-treated tubular cells stimulated renal fibroblasts, and this effect was suppressed by FGF2 neutralizing antibody and also by fgf2- or atg7-deletion in tubular cells. In human, compared with non-AKI, the renal biopsies from post-AKI patients had higher levels of autophagy and FGF2 in tubular cells, which showed significant correlations with renal fibrosis. These results indicate that persistent autophagy after AKI induces pro-fibrotic phenotype transformation in tubular cells leading to the expression and secretion of FGF2, which activates fibroblasts for renal fibrosis during maladaptive kidney repair.Abbreviations: 3-MA: 3-methyladnine; ACTA2/α-SMA: actin alpha 2, smooth muscle, aorta; ACTB/β-actin: actin, beta; AKI: acute kidney injury; ATG/Atg: autophagy related; BUN: blood urea nitrogen; CCN2/CTGF: cellular communication network factor 2; CDKN2A/p16: cyclin dependent kinase inhibitor 2A; CKD: chronic kidney disease; CM: conditioned medium; COL1A1: collagen, type I, alpha 1; COL4A1: collagen, type IV, alpha 1; CQ: chloroquine; ECM: extracellular matrix; eGFR: estimated glomerular filtration rate; ELISA: enzyme-linked immunosorbent assay; FGF2: fibroblast growth factor 2; FN1: fibronectin 1; FOXO3: forkhead box O3; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HAVCR1/KIM-1: hepatitis A virus cellular receptor 1; IHC: immunohistochemistry; IRI: ischemia-reperfusion injury; ISH: in situ hybridization; LTL: lotus tetragonolobus lectin; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; PDGFB: platelet derived growth factor, B polypeptide; PPIB/cyclophilin B: peptidylprolyl isomerase B; RT-qPCR: real time-quantitative PCR; SA-GLB1/β-gal: senescence-associated galactosidase, beta 1; SASP: senescence-associated secretory phenotype; sCr: serum creatinine; SQSTM1/p62: sequestosome 1; TASCC: TOR-autophagy spatial coupling compartment; TGFB1/TGF-β1: transforming growth factor, beta 1; VIM: vimentin.
Collapse
Affiliation(s)
- Man J. Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA,Research Department, Charlie Norwood VA Medical Center, Augusta, GA, USA,Man J. Livingston Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Shaoqun Shu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ze Li
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA,Research Department, Charlie Norwood VA Medical Center, Augusta, GA, USA,CONTACT Zheng Dong Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA30912, USA
| |
Collapse
|
44
|
Quimby JM, McLeland SM, Cianciolo RE, Lunn KF, Lulich JP, Erikson A, Barron LB. Frequency of histologic lesions in the kidneys of cats without kidney disease. J Feline Med Surg 2022; 24:e472-e480. [PMID: 36475921 PMCID: PMC10812332 DOI: 10.1177/1098612x221123768] [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] [Accepted: 08/12/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES In humans, renal aging is associated with an increased frequency of glomerulosclerosis, interstitial fibrosis, inflammation and tubular atrophy. The purpose of this study was to describe the frequency of renal histopathologic lesions in cats without kidney disease. METHODS A cross-sectional study of archival kidney tissue from 74 cats without kidney disease (serum creatinine <1.6 mg/dl; urine specific gravity >1.035) was carried out: 0-4 years (young, n = 18); 5-9 years (mature, n = 16); 10-14 years (senior, n = 34), 15+ years (geriatric, n = 6). Glomerulosclerosis, tubular atrophy, interstitial inflammation and fibrosis, and the presence or absence of lipid in the interstitium and tubules were scored by a pathologist masked to clinical data. Statistical analyses were performed as appropriate. RESULTS Geriatric cats had significantly more glomerulosclerosis than mature (P = 0.01) and young cats (P = 0.004). Senior cats had significantly more glomerulosclerosis than young cats (P = 0.006). Glomerulosclerosis was weakly positively correlated with age (r = 0.48; P <0.0001). Geriatric cats had significantly more tubular atrophy than mature (P = 0.02) and young cats (P <0.0001). Senior cats had significantly more tubular atrophy than young cats (P <0.0001). Geriatric cats had significantly more inflammation than senior cats (P = 0.02), mature cats (P = 0.01) and young cats (P <0.0001). Senior cats had significantly more inflammation than young cats (P = 0.004). Geriatric and senior cats had significantly more fibrosis than young cats (P = 0.01 and P = 0.04, respectively). Frequency of tubular lipid increased with age (young: 28%; mature: 56%; senior: 79%; geriatric: 100%) as did the frequency of interstitial lipid (young: 22%, mature: 56%, senior: 85%, geriatric: 100%). CONCLUSIONS AND RELEVANCE Evidence of renal aging exists in cats. These changes imply that the aging kidney may be more susceptible to injury and impaired healing.
Collapse
Affiliation(s)
- Jessica M Quimby
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Shannon M McLeland
- International Veterinary Renal Pathology Service, Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Rachel E Cianciolo
- International Veterinary Renal Pathology Service, Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Katharine F Lunn
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Jody P Lulich
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, MN, USA
| | - Andrea Erikson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Lara B Barron
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, MN, USA
| |
Collapse
|
45
|
Ciarambino T, Crispino P, Giordano M. Gender and Renal Insufficiency: Opportunities for Their Therapeutic Management? Cells 2022; 11:cells11233820. [PMID: 36497080 PMCID: PMC9740491 DOI: 10.3390/cells11233820] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is a major clinical problem associated with increased morbidity and mortality. Despite intensive research, the clinical outcome remains poor, and apart from supportive therapy, no other specific therapy exists. Furthermore, acute kidney injury increases the risk of developing chronic kidney disease (CKD) and end-stage renal disease. Acute tubular injury accounts for the most common intrinsic cause of AKI. The main site of injury is the proximal tubule due to its high workload and energy demand. Upon injury, an intratubular subpopulation of proximal epithelial cells proliferates and restores the tubular integrity. Nevertheless, despite its strong regenerative capacity, the kidney does not always achieve its former integrity and function and incomplete recovery leads to persistent and progressive CKD. Clinical and experimental data demonstrate sexual differences in renal anatomy, physiology, and susceptibility to renal diseases including but not limited to ischemia-reperfusion injury. Some data suggest the protective role of female sex hormones, whereas others highlight the detrimental effect of male hormones in renal ischemia-reperfusion injury. Although the important role of sex hormones is evident, the exact underlying mechanisms remain to be elucidated. This review focuses on collecting the current knowledge about sexual dimorphism in renal injury and opportunities for therapeutic manipulation, with a focus on resident renal progenitor stem cells as potential novel therapeutic strategies.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81031 Caserta, Italy
- Correspondence: (T.C.); (M.G.)
| | - Pietro Crispino
- Emergency Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy
| | - Mauro Giordano
- Department of Advanced Medical and Surgical Science, University of Campania, Luigi Vanvitelli, 80138 Naples, Italy
- Correspondence: (T.C.); (M.G.)
| |
Collapse
|
46
|
Protective Effects of Carnosol on Renal Interstitial Fibrosis in a Murine Model of Unilateral Ureteral Obstruction. Antioxidants (Basel) 2022; 11:antiox11122341. [PMID: 36552549 PMCID: PMC9774539 DOI: 10.3390/antiox11122341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Renal fibrosis is a common feature of chronic kidney disease and is a promising therapeutic target. However, there is still limited treatment for renal fibrosis, so the development of new anti-fibrotic agents is urgently needed. Accumulating evidence suggest that oxidative stress and endoplasmic reticulum (ER) stress play a critical role in renal fibrosis. Carnosol (CS) is a bioactive diterpene compound present in rosemary plants and has potent antioxidant and anti-inflammatory properties. In this study, we investigated the potential effects of CS on renal injury and fibrosis in a murine model of unilateral ureteral obstruction (UUO). Male C57BL/6J mice underwent sham or UUO surgery and received intraperitoneal injections of CS (50 mg/kg) daily for 8 consecutive days. CS improved renal function and ameliorated renal tubular injury and interstitial fibrosis in UUO mice. It suppressed oxidative injury by inhibiting pro-oxidant enzymes and activating antioxidant enzymes. Activation of ER stress was also attenuated by CS. In addition, CS inhibited apoptotic and necroptotic cell death in kidneys of UUO mice. Furthermore, cytokine production and immune cell infiltration were alleviated by CS. Taken together, these findings indicate that CS can attenuate renal injury and fibrosis in the UUO model.
Collapse
|
47
|
Hansson E, Wesseling C, Wegman D, Ekström U, Chavarria D, Glaser J, Jakobsson K. Point-of-care biomarkers for prediction of kidney function trajectory among sugarcane cutters: a comparative test accuracy study. BMJ Open 2022; 12:e060364. [PMID: 36400724 PMCID: PMC9677021 DOI: 10.1136/bmjopen-2021-060364] [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] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES Heat-stressed Mesoamerican workers, such as sugarcane cutters, suffer from high rates of chronic kidney disease of non-traditional origin (CKDnt). We aimed to identify easily available early markers of rapid kidney function decline in a population at high risk of CKDnt. DESIGN The accuracy of different biomarkers measured during harvest for prediction of cross-harvest kidney function decline were assessed in an exploratory study group, and the performance of the most promising biomarker was then assessed in an independent confirmation group. SETTING Male sugarcane cutters in El Salvador and Nicaragua. PARTICIPANTS 39 male Salvadoran sugarcane cutters sampled fortnightly at ≤9 occasions before and after work shift during harvest. 371 male Nicaraguan sugarcane cutters were sampled as part of routine monitoring during two harvests. Cutters worked at high physical intensity at wet-bulb globe temperatures mostly above 29°C for 6-8 hours per day 6 days a week during the 5-6 months harvest season. PRIMARY OUTCOMES Change in estimated glomerular filtration rate (CKD Epidemiology Collaboration) across the harvest season (ΔeGFRcross-harvest). RESULTS Dipstick leukocyturia after work shift in the El Salvadoran group was the most promising marker, explaining >25% of ΔeGFRcross-harvest variance at 8/9 occasions during harvest. Leukocyturia was associated with experiencing fever, little or dark urine, cramps, headache, dizziness and abdominal pain in the preceding 2-week period. Decreasing blood haemoglobin (Hb) and eGFR during harvest were also predictive of ΔeGFRcross-harvest. In the Nicaraguan confirmation dataset, those having ≥++ leukocyturia at any sampling during harvest had a 13 mL/min/1.73 m2 (95% CI 10 to 16 mL/min/1.73 m2) worse ΔeGFRcross-harvest than those without recorded leukocyturia. CONCLUSION Leukocyturia and Hb, both measurable with point-of-care methods, may be early indicators for kidney injury and risk for eGFR decline among heat-stressed male workers, thereby facilitating individual-level prevention and research aiming to understand the causes of CKDnt.
Collapse
Affiliation(s)
- Erik Hansson
- La Isla Network, Washington, District of Columbia, USA
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Catharina Wesseling
- La Isla Network, Washington, District of Columbia, USA
- Institute of Environmental Medicine, Unit of Occupational Medicine, Karolinska Institute, Stockholm, Sweden
| | - David Wegman
- La Isla Network, Washington, District of Columbia, USA
- University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Ulf Ekström
- La Isla Network, Washington, District of Columbia, USA
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Denis Chavarria
- Occupational Health Management, Ingenio San Antonio/Nicaragua Sugar Estates Limited, Chichigalpa, Nicaragua
| | - Jason Glaser
- La Isla Network, Washington, District of Columbia, USA
| | - Kristina Jakobsson
- La Isla Network, Washington, District of Columbia, USA
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Occupational and Environmental Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
48
|
Lausecker F, Lennon R, Randles MJ. The kidney matrisome in health, aging, and disease. Kidney Int 2022; 102:1000-1012. [PMID: 35870643 DOI: 10.1016/j.kint.2022.06.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 02/06/2023]
Abstract
Dysregulated extracellular matrix is the hallmark of fibrosis, and it has a profound impact on kidney function in disease. Furthermore, perturbation of matrix homeostasis is a feature of aging and is associated with declining kidney function. Understanding these dynamic processes, in the hope of developing therapies to combat matrix dysregulation, requires the integration of data acquired by both well-established and novel technologies. Owing to its complexity, the extracellular proteome, or matrisome, still holds many secrets and has great potential for the identification of clinical biomarkers and drug targets. The molecular resolution of matrix composition during aging and disease has been illuminated by cutting-edge mass spectrometry-based proteomics in recent years, but there remain key questions about the mechanisms that drive altered matrix composition. Basement membrane components are particularly important in the context of kidney function; and data from proteomic studies suggest that switches between basement membrane and interstitial matrix proteins are likely to contribute to organ dysfunction during aging and disease. Understanding the impact of such changes on physical properties of the matrix, and the subsequent cellular response to altered stiffness and viscoelasticity, is of critical importance. Likewise, the comparison of proteomic data sets from multiple organs is required to identify common matrix biomarkers and shared pathways for therapeutic intervention. Coupled with single-cell transcriptomics, there is the potential to identify the cellular origin of matrix changes, which could enable cell-targeted therapy. This review provides a contemporary perspective of the complex kidney matrisome and draws comparison to altered matrix in heart and liver disease.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Michael J Randles
- Chester Medical School, Faculty of Medicine and Life Sciences, University of Chester, Chester, UK.
| |
Collapse
|
49
|
Jang HS, Noh MR, Plumb T, Lee K, He JC, Ferrer FA, Padanilam BJ. Hepatic and proximal tubule angiotensinogen play distinct roles in kidney dysfunction, glomerular and tubular injury, and fibrosis progression. Am J Physiol Renal Physiol 2022; 323:F435-F446. [PMID: 35924445 PMCID: PMC9485008 DOI: 10.1152/ajprenal.00029.2022] [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: 02/09/2022] [Revised: 06/21/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Components of the renin-angiotensin system, including angiotensinogen (AGT), are critical contributors to chronic kidney disease (CKD) development and progression. However, the specific role of tissue-derived AGTs in CKD has not been fully understood. To define the contribution of liver versus kidney AGT in the CKD development, we performed 5/6 nephrectomy (Nx), an established CKD model, in wild-type (WT), proximal tubule (PT)- or liver-specific AGT knockout (KO) mice. Nx significantly elevated intrarenal AGT expression and elevated blood pressure (BP) in WT mice. The increase of intrarenal AGT protein was completely blocked in liver-specific AGT KO mice with BP reduction, suggesting a crucial role for liver AGT in BP regulation during CKD. Nx-induced glomerular and kidney injury and dysfunction, as well as fibrosis, were all attenuated to a greater extent in liver-specific AGT KO mice compared with PT-specific AGT KO and WT mice. However, the suppression of interstitial fibrosis in PT- and liver-specific AGT KO mouse kidneys was comparable. Our findings demonstrate that liver AGT acts as a critical contributor in driving glomerular and tubular injury, renal dysfunction, and fibrosis progression, whereas the role of PT AGT was limited to interstitial fibrosis progression in chronic renal insufficiency. Our results provide new insights for the development of tissue-targeted renin-angiotensin system intervention in the treatment of CKD.NEW & NOTEWORTHY Chronic kidney disease (CKD) is a major unmet medical need with no effective treatment. Current findings demonstrate that hepatic and proximal tubule angiotensinogen have distinct roles in tubular and glomerular injury, fibrogenesis, and renal dysfunction during CKD development. As renin-angiotensin system components, including angiotensinogen, are important targets for treating CKD in the clinic, the results from our study may be applied to developing better tissue-targeted treatment strategies for CKD and other fibroproliferative diseases.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mi Ra Noh
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Troy Plumb
- Division of Nephrology, Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fernando A Ferrer
- Department of Urology, Kravis Children's Hospital, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Babu J Padanilam
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
50
|
Tanemoto F, Nangaku M, Mimura I. Epigenetic memory contributing to the pathogenesis of AKI-to-CKD transition. Front Mol Biosci 2022; 9:1003227. [PMID: 36213117 PMCID: PMC9532834 DOI: 10.3389/fmolb.2022.1003227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022] Open
Abstract
Epigenetic memory, which refers to the ability of cells to retain and transmit epigenetic marks to their daughter cells, maintains unique gene expression patterns. Establishing programmed epigenetic memory at each stage of development is required for cell differentiation. Moreover, accumulating evidence shows that epigenetic memory acquired in response to environmental stimuli may be associated with diverse diseases. In the field of kidney diseases, the “memory” of acute kidney injury (AKI) leads to progression to chronic kidney disease (CKD); epidemiological studies show that patients who recover from AKI are at high risk of developing CKD. The underlying pathological processes include nephron loss, maladaptive epithelial repair, inflammation, and endothelial injury with vascular rarefaction. Further, epigenetic alterations may contribute as well to the pathophysiology of this AKI-to-CKD transition. Epigenetic changes induced by AKI, which can be recorded in cells, exert long-term effects as epigenetic memory. Considering the latest findings on the molecular basis of epigenetic memory and the pathophysiology of AKI-to-CKD transition, we propose here that epigenetic memory contributing to AKI-to-CKD transition can be classified according to the presence or absence of persistent changes in the associated regulation of gene expression, which we designate “driving” memory and “priming” memory, respectively. “Driving” memory, which persistently alters the regulation of gene expression, may contribute to disease progression by activating fibrogenic genes or inhibiting renoprotective genes. This process may be involved in generating the proinflammatory and profibrotic phenotypes of maladaptively repaired tubular cells after kidney injury. “Priming” memory is stored in seemingly successfully repaired tubular cells in the absence of detectable persistent phenotypic changes, which may enhance a subsequent transcriptional response to the second stimulus. This type of memory may contribute to AKI-to-CKD transition through the cumulative effects of enhanced expression of profibrotic genes required for wound repair after recurrent AKI. Further understanding of epigenetic memory will identify therapeutic targets of future epigenetic intervention to prevent AKI-to-CKD transition.
Collapse
|