1
|
Du Y, Wu M, Song S, Bian Y, Shi Y. TXNIP deficiency attenuates renal fibrosis by modulating mTORC1/TFEB-mediated autophagy in diabetic kidney disease. Ren Fail 2024; 46:2338933. [PMID: 38616177 PMCID: PMC11018024 DOI: 10.1080/0886022x.2024.2338933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 03/30/2024] [Indexed: 04/16/2024] Open
Abstract
Thioredoxin-interacting protein (TXNIP) is an important regulatory protein for thioredoxin (TRX) that elicits the generation of reactive oxygen species (ROS) by inhibiting the redox function of TRX. Abundant evidence suggests that TXNIP is involved in the fibrotic process of diabetic kidney disease (DKD). However, the potential mechanism of TXNIP in DKD is not yet well understood. In this study, we found that TXNIP knockout suppressed renal fibrosis and activation of mammalian target of rapamycin complex 1 (mTORC1) and restored transcription factor EB (TFEB) and autophagy activation in diabetic kidneys. Simultaneously, TXNIP interference inhibited epithelial-to-mesenchymal transformation (EMT), collagen I and fibronectin expression, and mTORC1 activation, increased TFEB nuclear translocation, and promoted autophagy restoration in HK-2 cells exposed to high glucose (HG). Rapamycin, an inhibitor of mTORC1, increased TFEB nuclear translocation and autophagy in HK-2 cells under HG conditions. Moreover, the TFEB activators, curcumin analog C1 and trehalose, effectively restored HG-induced autophagy, and abrogated HG-induced EMT and collagen I and fibronectin expression in HK-2 cells. Taken together, these findings suggest that TXNIP deficiency ameliorates renal fibrosis by regulating mTORC1/TFEB-mediated autophagy in diabetic kidney diseases.
Collapse
Affiliation(s)
- Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Kajimoto E, Nagasu H, Takasu M, Kishi S, Wada M, Tatsugawa R, Hirano A, Iwakura T, Umeno R, Wada Y, Itano S, Kadoya H, Kidokoro K, Sasaki T, Kashihara N. ASK-1 activation exacerbates kidney dysfunction via increment of glomerular permeability and accelerates cellular aging in diabetic kidney disease model mice. Sci Rep 2024; 14:26438. [PMID: 39488570 DOI: 10.1038/s41598-024-77577-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
Diabetic kidney disease (DKD) is a major disease characterized by early albuminuria and heightened risk of renal deterioration. Increased reactive oxygen species (ROS) production, especially in glomeruli, plays an important role in the progression of DKD. ROS also cause activation of Apoptosis signal-regulating kinase 1 (ASK-1), which is implicated in various organ injuries. However, the detailed mechanisms remain unclear. This study investigates ASK-1 activation in advanced DKD and its underlying mechanisms using GS442172, an ASK-1 inhibitor. In the DKD mouse model, activation of ASK-1 was observed. Although inhibition of ASK-1 activation improved hyperpermeability in glomerular endothelial cells. ASK-1 inhibition significantly reduced glomerular injury and albuminuria, while also attenuating tubular damage and interstitial fibrosis. RNA-seq analysis revealed an aging phenotype associated with ASK-1 activation in DKD. In vitro experiments demonstrated ASK-1 activation-induced cellular senescence in tubular cells via redox signaling. These results suggested that the critical role of ASK-1 activation in DKD pathogenesis, implicating glomerular injury, tubular damage, and cellular senescence. ASK-1 inhibitors are promising therapeutic strategies to mitigate the progression of DKD.
Collapse
Affiliation(s)
- Eriko Kajimoto
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Masanobu Takasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Seiji Kishi
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Masafumi Wada
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Rie Tatsugawa
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Akira Hirano
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Tsukasa Iwakura
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Reina Umeno
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Yoshihisa Wada
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Seiji Itano
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Hiroyuki Kadoya
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
- Department of General Geriatric Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kengo Kidokoro
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Tamaki Sasaki
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | | |
Collapse
|
3
|
Yang K, Liang W, Hu H, Zhang Z, Hao Y, Song Z, Yang L, Hu J, Chen Z, Ding G. ESRRA modulation by empagliflozin mitigates diabetic tubular injury via mitochondrial restoration. Cell Signal 2024; 122:111308. [PMID: 39059756 DOI: 10.1016/j.cellsig.2024.111308] [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: 02/19/2024] [Revised: 07/09/2024] [Accepted: 07/21/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND The protection of the diabetic kidney by Empagliflozin (EMPA) is attributed to its interaction with the sodium glucose cotransporter 2 located on proximal tubular epithelial cells (PTECs). Estrogen-related receptor α (ESRRA), known for its high expression in PTECs and association with mitochondrial biogenesis, plays a crucial role in this process. This study aimed to explore the impact of ESRRA on mitochondrial mass in diabetic tubular injury and elucidate the mechanism underlying the protective effects of EMPA. METHODS Mitochondrial changes in PTECs of 16-week-old diabetic mice were assessed using transmission electron microscopy (TEM) and RNA-sequences. In vivo, EMPA administration was carried out in db/db mice for 8 weeks, while in vitro experiments involved modifying ESRRA expression in HK2 cells using pcDNA-ESRRA or EMPA. RESULTS Evaluation through TEM revealed reduced mitochondrial mass and swollen mitochondria in PTECs, whereas no significant changes were observed under light microscopy. Analysis of RNA-sequences identified 110 downregulated genes, including Esrra, associated with mitochondrial function. Notably, ESRRA overexpression rescued the loss of mitochondrial mass induced by high glucose (HG) in HK2 cells. EMPA treatment ameliorated the ultrastructural alterations and mitigated the downregulation of ESRRA both in db/db mice and HG-treated HK2 cells. CONCLUSION The diminished expression of ESRRA is implicated in the decline of mitochondrial mass in PTECs during the early stages of diabetes, highlighting it as a key target of EMPA for preventing the progression of diabetic kidney injury.
Collapse
Affiliation(s)
- Keju Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhixia Song
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Lin Yang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
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
|
5
|
Tao Y, Lacko AG, Sabnis NA, Das‐Earl P, Ibrahim D, Crowe N, Zhou Z, Cunningham M, Castillo A, Ma R. Reconstituted HDL ameliorated renal injury of diabetic kidney disease in mice. Physiol Rep 2024; 12:e16179. [PMID: 39107084 PMCID: PMC11303015 DOI: 10.14814/phy2.16179] [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: 03/06/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/09/2024] Open
Abstract
Diabetic kidney disease (DKD) is a devastating kidney disease and lacks effective therapeutic interventions. The present study was aimed to determine whether reconstituted high-density lipoprotein (rHDL) ameliorated renal injury in eNOS-/- dbdb mice, a mouse model of DKD. Three groups of mice, wild type C57BLKS/J (non-diabetes), eNOS-/- dbdb (diabetes), and eNOS-/- dbdb treated with rHDL (diabetes+rHDL) with both males and females were used. The rHDL nanoparticles were administered to eNOS-/- dbdb mice at Week 16 at 5 μg/g body weight in ~100 μL of saline solution twice per week for 4 weeks via retroorbital injection. We found that rHDL treatment significantly blunted progression of albuminuria and GFR decline observed in DKD mice. Histological examinations showed that the rHDLs significantly alleviated glomerular injury and renal fibrosis, and inhibited podocyte loss. Western blots and immunohistochemical examinations showed that increased protein abundances of fibronectin and collagen IV in the renal cortex of eNOS-/- dbdb mice were significantly reduced by the rHDLs. Taken together, the present study suggests a renoprotective effect of rHDLs on DKD.
Collapse
Affiliation(s)
- Yu Tao
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Andras G. Lacko
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Nirupama A. Sabnis
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Paromita Das‐Earl
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Deena Ibrahim
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Nicole Crowe
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Zhengyang Zhou
- Department of Population and Community HealthUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Mark Cunningham
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Angie Castillo
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Rong Ma
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
6
|
Liu X, Li MH, Zhao YY, Xie YL, Yu X, Chen YJ, Li P, Zhang WF, Zhu TT. LncRNA H19 deficiency protects against the structural damage of glomerular endothelium in diabetic nephropathy via Akt/eNOS pathway. Arch Physiol Biochem 2024; 130:401-410. [PMID: 35867533 DOI: 10.1080/13813455.2022.2102655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022]
Abstract
Objective: This study aimed to investigate the functions of lncRNA H19 on glomerular endothelial structural damage of diabetic nephropathy (DN).Materials and Methods: Rats were fed a high sugar and fat high feed die, and intraperitoneally administrated with streptozotocin (30 mg/kg) to induce DN model. Meanwile, rat glomerular endothelial cells (rGEnCs) were treated with high a level of glucose (HG, 30 mM glucose)to induce structural damage.Results: Our results showed that H19 level was drastically increased in diabetic glomeruli and high-glucose (HG)-stimulated rat glomerular endothelial cells (rGEnCs). Deficiency of H19 ameliorated microalbumin, creatinine, BUN, and histopathological alterations in diabetic rats. In addition, H19 deficiency significantly attenuated the damage of endothelial structure by upregulating the expression of junction proteins ZO-1 and Occludin, glycolcalyx protein Syndecan-1, and endothelial activation marker sVCAM-1 and sICAM-1 in diabetic rats. The in vitro results also showed that H19-siRNA alleviated glycocalyx shedding, tight junctions damage, and endothelial activation in HG-stimulated rGEnCs. Moreover, H19 deficiency significantly enhanced the expression of p-Akt and p-eNOS and NO concentration in vitro and in vivo. Pre-treatment with Akt inhibitor LY294002 abrogated these favourable effects mediated by H19 deficiency.Discussion and Conclusion: These results indicate that H19 deficiency could mitigate the structural damage of glomerular endothelium in DN via activating Akt/eNOS pathway.
Collapse
Affiliation(s)
- Xu Liu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ming-Hui Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yun-Yun Zhao
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu-Liang Xie
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xin Yu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu-Jing Chen
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Wei-Fang Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Tian-Tian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
- Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| |
Collapse
|
7
|
Min L, Zhong F, Gu L, Lee K, He JC. Krüppel-like factor 2 is an endoprotective transcription factor in diabetic kidney disease. Am J Physiol Cell Physiol 2024; 327:C477-C486. [PMID: 38981608 DOI: 10.1152/ajpcell.00222.2024] [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: 04/11/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes, and glomerular endothelial cell (GEC) dysfunction is a key driver of DKD pathogenesis. Krüppel-like factor 2 (KLF2), a shear stress-induced transcription factor, is among the highly regulated genes in early DKD. In the kidney, KLF2 expression is mostly restricted to endothelial cells, but its expression is also found in immune cell subsets. KLF2 expression is upregulated in response to increased shear stress by the activation of mechanosensory receptors but suppressed by inflammatory cytokines, both of which characterize the early diabetic kidney milieu. KLF2 expression is reduced in progressive DKD and hypertensive nephropathy in humans and mice, likely due to high glucose and inflammatory cytokines such as TNF-α. However, KLF2 expression is increased in glomerular hyperfiltration-induced shear stress without metabolic dysregulation, such as in settings of unilateral nephrectomy. Lower KLF2 expression is associated with CKD progression in patients with unilateral nephrectomy, consistent with its endoprotective role. KLF2 confers endoprotection by inhibition of inflammation, thrombotic activation, and angiogenesis, and thus KLF2 is considered a protective factor for cardiovascular disease (CVD). Based on similar mechanisms, KLF2 also exhibits renoprotection, and its reduced expression in endothelial cells worsens glomerular injury and albuminuria in settings of diabetes or unilateral nephrectomy. Thus KLF2 confers endoprotective effects in both CVD and DKD, and its activators could potentially be developed as a novel class of drugs for cardiorenal protection in diabetic patients.
Collapse
Affiliation(s)
- Lulin Min
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Fang Zhong
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kyung Lee
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - John Cijiang He
- Department of Medicine/Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, New York, United States
| |
Collapse
|
8
|
Gaudet A, Zheng X, Kambham N, Bhalla V. Esm-1 mediates transcriptional polarization associated with diabetic kidney disease. Am J Physiol Renal Physiol 2024; 326:F1016-F1031. [PMID: 38601985 PMCID: PMC11386982 DOI: 10.1152/ajprenal.00419.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/20/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
Esm-1, endothelial cell-specific molecule-1, is a susceptibility gene for diabetic kidney disease (DKD) and is a secreted proteoglycan, with notable expression in kidney, which attenuates inflammation and albuminuria. However, little is known about Esm1 expression in mature tissues in the presence or absence of diabetes. We utilized publicly available single-cell RNA sequencing data to characterize Esm1 expression in 27,786 renal endothelial cells (RECs) obtained from three mouse and four human databases. We validated our findings using bulk transcriptome data from 20 healthy subjects and 41 patients with DKD and using RNAscope. In both mice and humans, Esm1 is expressed in a subset of all REC types and represents a minority of glomerular RECs. In patients, Esm1(+) cells exhibit conserved enrichment for blood vessel development genes. With diabetes, these cells are fewer in number and shift expression toward chemotaxis pathways. Esm1 correlates with a majority of genes within these pathways, delineating a glomerular transcriptional polarization reflected by the magnitude of Esm1 deficiency. Diabetes correlates with lower Esm1 expression and with changes in the functional characterization of Esm1(+) cells. Thus, Esm1 appears to be a marker for glomerular transcriptional polarization in DKD.NEW & NOTEWORTHY Esm-1 is primarily expressed in glomerular endothelium in humans. Cells expressing Esm1 exhibit a high degree of conservation in the enrichment of genes related to blood vessel development. In the context of diabetes, these cells are reduced in number and show a significant transcriptional shift toward the chemotaxis pathway. In diabetes, there is a transcriptional polarization in the glomerulus that is reflected by the degree of Esm1 deficiency.
Collapse
Affiliation(s)
- Alexandre Gaudet
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Xiaoyi Zheng
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States
| | - Neeraja Kambham
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States
| |
Collapse
|
9
|
Juin SK, Pushpakumar S, Sen U. Nimbidiol protects from renal injury by alleviating redox imbalance in diabetic mice. Front Pharmacol 2024; 15:1369408. [PMID: 38835661 PMCID: PMC11148448 DOI: 10.3389/fphar.2024.1369408] [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: 01/12/2024] [Accepted: 04/17/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction Chronic hyperglycemia-induced oxidative stress plays a crucial role in the development of diabetic nephropathy (DN). Moreover, adverse extracellular matrix (ECM) accumulation elevates renal resistive index leading to progressive worsening of the pathology in DN. Nimbidiol is an alpha-glucosidase inhibitor, isolated from the medicinal plant, 'neem' (Azadirachta indica) and reported as a promising anti-diabetic compound. Previously, a myriad of studies demonstrated an anti-oxidative property of a broad-spectrum neem-extracts in various diseases including diabetes. Our recent study has shown that Nimbidiol protects diabetic mice from fibrotic renal dysfunction in part by mitigating adverse ECM accumulation. However, the precise mechanism remains poorly understood. Methods The present study aimed to investigate whether Nimbidiol ameliorates renal injury by reducing oxidative stress in type-1 diabetes. To test the hypothesis, wild-type (C57BL/6J) and diabetic Akita (C57BL/6-Ins2Akita/J) mice aged 10-14 weeks were used to treat with saline or Nimbidiol (400 μg kg-1 day-1) for 8 weeks. Results Diabetic mice showed elevated blood pressure, increased renal resistive index, and decreased renal vasculature compared to wild-type control. In diabetic kidney, reactive oxygen species and the expression levels of 4HNE, p22phox, Nox4, and ROMO1 were increased while GSH: GSSG, and the expression levels of SOD-1, SOD-2, and catalase were decreased. Further, eNOS, ACE2, Sirt1 and IL-10 were found to be downregulated while iNOS and IL-17 were upregulated in diabetic kidney. The changes were accompanied by elevated expression of the renal injury markers viz., lipocalin-2 and KIM-1 in diabetic kidney. Moreover, an upregulation of p-NF-κB and a downregulation of IkBα were observed in diabetic kidney compared to the control. Nimbidiol ameliorated these pathological changes in diabetic mice. Conclusion Altogether, the data of our study suggest that oxidative stress largely contributes to the diabetic renal injury, and Nimbidiol mitigates redox imbalance and thereby protects kidney in part by inhibiting NF-κB signaling pathway in type-1 diabetes.
Collapse
Affiliation(s)
- Subir Kumar Juin
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
10
|
Wang N, Zhang C. Oxidative Stress: A Culprit in the Progression of Diabetic Kidney Disease. Antioxidants (Basel) 2024; 13:455. [PMID: 38671903 PMCID: PMC11047699 DOI: 10.3390/antiox13040455] [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: 02/27/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic kidney disease (DKD) is the principal culprit behind chronic kidney disease (CKD), ultimately developing end-stage renal disease (ESRD) and necessitating costly dialysis or kidney transplantation. The limited therapeutic efficiency among individuals with DKD is a result of our finite understanding of its pathogenesis. DKD is the result of complex interactions between various factors. Oxidative stress is a fundamental factor that can establish a link between hyperglycemia and the vascular complications frequently encountered in diabetes, particularly DKD. It is crucial to recognize the essential and integral role of oxidative stress in the development of diabetic vascular complications, particularly DKD. Hyperglycemia is the primary culprit that can trigger an upsurge in the production of reactive oxygen species (ROS), ultimately sparking oxidative stress. The main endogenous sources of ROS include mitochondrial ROS production, NADPH oxidases (Nox), uncoupled endothelial nitric oxide synthase (eNOS), xanthine oxidase (XO), cytochrome P450 (CYP450), and lipoxygenase. Under persistent high glucose levels, immune cells, the complement system, advanced glycation end products (AGEs), protein kinase C (PKC), polyol pathway, and the hexosamine pathway are activated. Consequently, the oxidant-antioxidant balance within the body is disrupted, which triggers a series of reactions in various downstream pathways, including phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor beta/p38-mitogen-activated protein kinase (TGF-β/p38-MAPK), nuclear factor kappa B (NF-κB), adenosine monophosphate-activated protein kinase (AMPK), and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. The disease might persist even if strict glucose control is achieved, which can be attributed to epigenetic modifications. The treatment of DKD remains an unresolved issue. Therefore, reducing ROS is an intriguing therapeutic target. The clinical trials have shown that bardoxolone methyl, a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, blood glucose-lowering drugs, such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists can effectively slow down the progression of DKD by reducing oxidative stress. Other antioxidants, including vitamins, lipoic acid, Nox inhibitors, epigenetic regulators, and complement inhibitors, present a promising therapeutic option for the treatment of DKD. In this review, we conduct a thorough assessment of both preclinical studies and current findings from clinical studies that focus on targeted interventions aimed at manipulating these pathways. We aim to provide a comprehensive overview of the current state of research in this area and identify key areas for future exploration.
Collapse
Affiliation(s)
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
11
|
Yu MG, Gordin D, Fu J, Park K, Li Q, King GL. Protective Factors and the Pathogenesis of Complications in Diabetes. Endocr Rev 2024; 45:227-252. [PMID: 37638875 PMCID: PMC10911956 DOI: 10.1210/endrev/bnad030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/13/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Chronic complications of diabetes are due to myriad disorders of numerous metabolic pathways that are responsible for most of the morbidity and mortality associated with the disease. Traditionally, diabetes complications are divided into those of microvascular and macrovascular origin. We suggest revising this antiquated classification into diabetes complications of vascular, parenchymal, and hybrid (both vascular and parenchymal) tissue origin, since the profile of diabetes complications ranges from those involving only vascular tissues to those involving mostly parenchymal organs. A major paradigm shift has occurred in recent years regarding the pathogenesis of diabetes complications, in which the focus has shifted from studies on risks to those on the interplay between risk and protective factors. While risk factors are clearly important for the development of chronic complications in diabetes, recent studies have established that protective factors are equally significant in modulating the development and severity of diabetes complications. These protective responses may help explain the differential severity of complications, and even the lack of pathologies, in some tissues. Nevertheless, despite the growing number of studies on this field, comprehensive reviews on protective factors and their mechanisms of action are not available. This review thus focused on the clinical, biochemical, and molecular mechanisms that support the idea of endogenous protective factors, and their roles in the initiation and progression of chronic complications in diabetes. In addition, this review also aimed to identify the main needs of this field for future studies.
Collapse
Affiliation(s)
- Marc Gregory Yu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel Gordin
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
- Department of Nephrology, University of Helsinki and Helsinki University Central Hospital, Stenbäckinkatu 9, FI-00029 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Jialin Fu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Kyoungmin Park
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Qian Li
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - George Liang King
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
12
|
Tomita-Yagi A, Ozeki-Okuno N, Watanabe-Uehara N, Komaki K, Umehara M, Sawada-Yamauchi H, Minamida A, Sunahara Y, Matoba Y, Nakamura I, Nakata T, Nakai K, Ida T, Yamashita N, Kamezaki M, Kirita Y, Taniguchi T, Konishi E, Matoba S, Tamagaki K, Kusaba T. The importance of proinflammatory failed-repair tubular epithelia as a predictor of diabetic kidney disease progression. iScience 2024; 27:109020. [PMID: 38357667 PMCID: PMC10865398 DOI: 10.1016/j.isci.2024.109020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/17/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
The immense public health burden of diabetic kidney disease (DKD) has led to an increase in research on the pathophysiology of advanced DKD. The present study focused on the significance of proinflammatory vascular cell adhesion molecule 1 (VCAM1)+ tubules in DKD progression. A retrospective cohort study of DKD patients showed that the percentage of VCAM1+ tubules in kidney samples was correlated with poor renal outcomes. We established an advanced DKD model by partial resection of the kidneys of db/db mice and demonstrated that it closely resembled the human advanced DKD phenotype, with tissue hypoxia, tubular DNA damage, tissue inflammation, and high tubular VCAM1 expression. Luseogliflozin ameliorated tissue hypoxia and proinflammatory responses, including VCAM1+ expression, in tubules. These findings suggest the potential of tubular VCAM1 as a histological marker for poor DKD outcomes. SGLT2 inhibitors may attenuate tissue hypoxia and subsequent tissue inflammation in advanced DKD, thereby ameliorating tubular injury.
Collapse
Affiliation(s)
- Aya Tomita-Yagi
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsuko Ozeki-Okuno
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriko Watanabe-Uehara
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazumi Komaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Minato Umehara
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroko Sawada-Yamauchi
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Minamida
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuto Sunahara
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yayoi Matoba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Itaru Nakamura
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Nakata
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kunihiro Nakai
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoharu Ida
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Noriyuki Yamashita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michitsugu Kamezaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuhei Kirita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuya Taniguchi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiichi Tamagaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Kanoo S, Goodluck H, Kim YC, Garrido AN, Crespo-Masip M, Lopez N, Zhang H, Gonzalez-Villalobos RA, Ma LJ, Vallon V. Deletion, but Not Heterozygosity, of eNOS Raises Blood Pressure and Aggravates Nephropathy in BTBR ob/ob Mice. Nephron Clin Pract 2024; 148:631-642. [PMID: 38301618 PMCID: PMC11291698 DOI: 10.1159/000536522] [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: 08/17/2023] [Accepted: 01/20/2024] [Indexed: 02/03/2024] Open
Abstract
INTRODUCTION ob/ob mice are a leptin-deficient type 2 diabetes mellitus model, which, on a BTBR background, mimics the glomerular pathophysiology of diabetic nephropathy (DN). Since leptin deficiency reduces blood pressure (BP) and endothelial nitric oxide synthase (eNOS) lowers BP and is kidney protective, we attempted to develop a more robust DN model by introducing eNOS deficiency in BTBR ob/ob mice. METHODS Six experimental groups included littermate male and female BTBR ob/ob or wild-type for ob (control) as well as wild-type (WT), heterozygote (HET), or knockout (KO) for eNOS. Systolic BP (by automated tail-cuff) and GFR (by FITC-sinistrin plasma kinetics) were determined in awake mice at 27-30 weeks of age, followed by molecular and histological kidney analyses. RESULTS Male and female ob/ob WT presented hyperglycemia and larger body and kidney weight, GFR, glomerular injury, and urine albumin to creatinine ratio (UACR) despite modestly lower BP versus control WT. These effects were associated with a higher tubular injury score and renal mRNA expression of NGAL only in males, whereas female ob/ob WT unexpectedly had lower KIM-1 and COL1A1 expression versus control WT, indicating sex differences. HET for eNOS did not consistently alter BP or renal outcome in control or ob/ob. In comparison, eNOS KO increased BP (15-25 mm Hg) and worsened renal markers of injury, inflammation and fibrosis, GFR, UACR, and survival rates, as observed in control and, more pronouncedly, in ob/ob mice and independent of sex. CONCLUSIONS Deletion, but not heterozygosity, of eNOS raises blood pressure and aggravates nephropathy in BTBR ob/ob mice.
Collapse
Affiliation(s)
- Sadhana Kanoo
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Helen Goodluck
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Young Chul Kim
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Aleix Navarro Garrido
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Maria Crespo-Masip
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Natalia Lopez
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | - Haiyan Zhang
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| | | | - Li-Jun Ma
- CVMR&PH Discovery, Janssen Research and Development, LLC. Cambridge, MA and Spring House, PA
| | - Volker Vallon
- Department of Medicine, University of California San Diego, La Jolla, USA & VA San Diego Healthcare System, San Diego, USA
| |
Collapse
|
14
|
Al Tuhaifi T, Zhong J, Yang HC, Fogo AB. Effects of Dipeptidyl Peptidase-4 Inhibitor and Angiotensin-Converting Enzyme Inhibitor on Experimental Diabetic Kidney Disease. J Transl Med 2024; 104:100305. [PMID: 38109999 PMCID: PMC10922867 DOI: 10.1016/j.labinv.2023.100305] [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: 02/11/2023] [Revised: 11/08/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease in the United States and worldwide. Proteinuria is a major marker of the severity of injury. Dipeptidyl peptidase-4 inhibitor (DPP-4I) increases incretin-related insulin production and is, therefore, used to treat diabetes. We investigated whether DPP4I could have direct effect on kidney independent of its hypoglycemic activity. We, therefore, tested the effects of DPP4I with or without angiotensin-converting enzyme inhibitor (ACEI) on the progression of diabetic nephropathy and albuminuria in a murine model of DKD. eNOS-/-db/db mice were randomized to the following groups at age 10 weeks and treated until sacrifice: baseline (sacrificed at week 10), untreated control, ACEI, DPP4I, and combination of DPP4I and ACEI (Combo, sacrificed at week 18). Systemic parameters and urine albumin-creatinine ratio were assessed at baseline, weeks 14, and 18. Kidney morphology, glomerular filtration rate (GFR), WT-1, a marker for differentiated podocytes, podoplanin, a marker of foot process integrity, glomerular collagen IV, and alpha-smooth muscle actin were assessed at the end of the study. All mice had hyperglycemia and proteinuria at study entry at week 10. Untreated control mice had increased albuminuria, progression of glomerular injury, and reduced GFR at week 18 compared with baseline. DPP4I alone reduced blood glucose and kidney DPP-4 activity but failed to protect against kidney injury compared with untreated control. ACEI alone and combination groups showed significantly reduced albuminuria and glomerular injury, and maintained GFR and WT-1+ cells. Only the combination group had significantly less glomerular collagen IV deposition and more podoplanin preservation than the untreated control. DPP-4I alone does not decrease the progression of kidney injury in the eNOS-/-db/db mouse model, suggesting that targeting only hyperglycemia is not an optimal treatment strategy for DKD. Combined DPP-4I with ACEI added more benefit to reducing the glomerular matrix.
Collapse
Affiliation(s)
- Tareq Al Tuhaifi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Nephrology Clinical Trials Center, Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jianyong Zhong
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hai-Chun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
15
|
Taguchi S, Azushima K, Yamaji T, Suzuki T, Abe E, Tanaka S, Hirota K, Tsukamoto S, Morita R, Kobayashi R, Kinguchi S, Yamashita A, Wakui H, Tamura K. Angiotensin II type 1 receptor-associated protein deletion combined with angiotensin II stimulation accelerates the development of diabetic kidney disease in mice on a C57BL/6 strain. Hypertens Res 2024; 47:55-66. [PMID: 37957242 DOI: 10.1038/s41440-023-01496-4] [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: 07/12/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
The progress in the research field of diabetic kidney disease (DKD) has been disturbed by the lack of reliable animal models. Angiotensin II (Ang II) type 1 receptor (AT1R)-associated protein (ATRAP) promotes internalization of AT1R and selectively inhibits pathological AT1R signaling. In this study, we investigated whether overactivation of the renin-angiotensin system (RAS) through a combination of ATRAP deletion with Ang II stimulation developed a progressive DKD model in C57BL/6 mice, which are resistant to the development of kidney injury. Eight-week-old male systemic ATRAP-knockout mice on the C57BL/6 strain (KO) and their littermate wild-type mice (Ctrl) were divided into five groups: 1) Ctrl, 2) Ctrl-streptozotocin (STZ), 3) KO-STZ, 4) Ctrl-STZ-Ang II, and 5) KO-STZ-Ang II. Ang II was administered for 6 weeks from 4 weeks after STZ administration. At 10 weeks after STZ administration, mice were euthanized to evaluate kidney injuries. Neither ATRAP deletion alone nor Ang II stimulation alone developed a progressive DKD model in STZ-induced diabetic C57BL/6 mice. However, a combination of ATRAP deletion with Ang II stimulation accelerated the development of DKD as manifested by overt albuminuria, glomerular hypertrophy, podocyte loss, mesangial expansion, kidney interstitial fibrosis and functional insufficiency, concomitant with increased angiotensinogen and AT1R expression in the kidneys. In STZ-induced diabetic C57BL/6 mice that are resistant to the development of kidney injury, the combination of ATRAP deletion and Ang II stimulation accelerates the development of DKD, which may be associated with intrarenal RAS overactivation.
Collapse
Affiliation(s)
- Shinya Taguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Takahiro Yamaji
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Toru Suzuki
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Eriko Abe
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shohei Tanaka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keigo Hirota
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryutaro Morita
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Kobayashi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akio Yamashita
- Department of Investigative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
16
|
Wu J, Wang Z, Cai M, Wang X, Lo B, Li Q, He JC, Lee K, Fu J. GPR56 Promotes Diabetic Kidney Disease Through eNOS Regulation in Glomerular Endothelial Cells. Diabetes 2023; 72:1652-1663. [PMID: 37579299 PMCID: PMC10588296 DOI: 10.2337/db23-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Although glomerular endothelial dysfunction is well recognized as contributing to the pathogenesis of diabetic kidney disease (DKD), the molecular pathways contributing to DKD pathogenesis in glomerular endothelial cells (GECs) are only partially understood. To uncover pathways that are differentially regulated in early DKD that may contribute to disease pathogenesis, we recently conducted a transcriptomic analysis of isolated GECs from diabetic NOS3-null mice. The analysis identified several potential mediators of early DKD pathogenesis, one of which encoded an adhesion G protein-coupled receptor-56 (GPR56), also known as ADGRG1. Enhanced glomerular expression of GPR56 was observed in human diabetic kidneys, which was negatively associated with kidney function. Using cultured mouse GECs, we observed that GPR56 expression was induced with exposure to advanced glycation end products, as well as in high-glucose conditions, and its overexpression resulted in decreased phosphorylation and expression of endothelial nitric oxide synthase (eNOS). This effect on eNOS by GPR56 was mediated by coupling of Gα12/13-RhoA pathway activation and Gαi-mediated cAMP/PKA pathway inhibition. The loss of GPR56 in mice led to a significant reduction in diabetes-induced albuminuria and glomerular injury, which was associated with reduced oxidative stress and restoration of eNOS expression in GECs. These findings suggest that GPR56 promotes DKD progression mediated, in part, through enhancing glomerular endothelial injury and dysfunction. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jinshan Wu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihong Wang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minchao Cai
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xuan Wang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Benjamin Lo
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
- Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, Bronx, NY
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jia Fu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
17
|
Patidar K, Versypt ANF. Logic-Based Modeling of Inflammatory Macrophage Crosstalk with Glomerular Endothelial Cells in Diabetic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535594. [PMID: 37066138 PMCID: PMC10104015 DOI: 10.1101/2023.04.04.535594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Diabetic kidney disease is a complication in 1 out of 3 patients with diabetes. Aberrant glucose metabolism in diabetes leads to an immune response causing inflammation and to structural and functional damage in the glomerular cells of the kidney. Complex cellular signaling lies at the core of metabolic and functional derangement. Unfortunately, the mechanism underlying the role of inflammation in glomerular endothelial cell dysfunction during diabetic kidney disease is not fully understood. Computational models in systems biology allow the integration of experimental evidence and cellular signaling networks to understand mechanisms involved in disease progression. We built a logic-based ordinary differential equations model to study macrophage-dependent inflammation in glomerular endothelial cells during diabetic kidney disease progression. We studied the crosstalk between macrophages and glomerular endothelial cells in the kidney using a protein signaling network stimulated with glucose and lipopolysaccharide. The network and model were built using the open-source software package Netflux. This modeling approach overcomes the complexity of studying network models and the need for extensive mechanistic details. The model simulations were fitted and validated against available biochemical data from in vitro experiments. The model identified mechanisms responsible for dysregulated signaling in macrophages and glomerular endothelial cells during diabetic kidney disease. In addition, we investigated the influence of signaling interactions and species that on glomerular endothelial cell morphology through selective knockdown and downregulation. We found that partial knockdown of VEGF receptor 1, PLC-γ, adherens junction proteins, and calcium partially recovered the endothelial cell fenestration size. Our model findings contribute to understanding signaling and molecular perturbations that affect the glomerular endothelial cells in the early stage of diabetic kidney disease.
Collapse
|
18
|
Lu J, Li XQ, Chen PP, Zhang JX, Liu L, Wang GH, Liu XQ, Jiang TT, Wang MY, Liu WT, Ruan XZ, Ma KL. Activation of acetyl-CoA synthetase 2 mediates kidney injury in diabetic nephropathy. JCI Insight 2023; 8:e165817. [PMID: 37870960 PMCID: PMC10619493 DOI: 10.1172/jci.insight.165817] [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: 10/06/2022] [Accepted: 09/05/2023] [Indexed: 10/25/2023] Open
Abstract
Albuminuria and podocyte injury are the key cellular events in the progression of diabetic nephropathy (DN). Acetyl-CoA synthetase 2 (ACSS2) is a nucleocytosolic enzyme responsible for the regulation of metabolic homeostasis in mammalian cells. This study aimed to investigate the possible roles of ACSS2 in kidney injury in DN. We constructed an ACSS2-deleted mouse model to investigate the role of ACSS2 in podocyte dysfunction and kidney injury in diabetic mouse models. In vitro, podocytes were chosen and transfected with ACSS2 siRNA and ACSS2 inhibitor and treated with high glucose. We found that ACSS2 expression was significantly elevated in the podocytes of patients with DN and diabetic mice. ACSS2 upregulation promoted phenotype transformation and inflammatory cytokine expression while inhibiting podocytes' autophagy. Conversely, ACSS2 inhibition improved autophagy and alleviated podocyte injury. Furthermore, ACSS2 epigenetically activated raptor expression by histone H3K9 acetylation, promoting activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway. Pharmacological inhibition or genetic depletion of ACSS2 in the streptozotocin-induced diabetic mouse model greatly ameliorated kidney injury and podocyte dysfunction. To conclude, ACSS2 activation promoted podocyte injury in DN by raptor/mTORC1-mediated autophagy inhibition.
Collapse
Affiliation(s)
- Jian Lu
- Department of Nephrology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xue Qi Li
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Pei Pei Chen
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jia Xiu Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Liang Liu
- Department of Intensive Care Unit, People’s Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Gui Hua Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiao Qi Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ting Ting Jiang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Meng Ying Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wen Tao Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiong Zhong Ruan
- John Moorhead Research Laboratory, Department of Renal Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Kun Ling Ma
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Chung JO, Park SY, Cho DH, Chung DJ, Chung MY. Relationship between plasma leucine-rich α-2-glycoprotein 1 and urinary albumin excretion in patients with type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1232021. [PMID: 37916147 PMCID: PMC10617030 DOI: 10.3389/fendo.2023.1232021] [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: 05/31/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023] Open
Abstract
Aims To explore the relationship between plasma leucine-rich α-2-glycoprotein 1 (LRG1) level and the degree of urinary albumin excretion in patients with type 2 diabetes. Methods We evaluated 332 patients with type 2 diabetes in a cross-sectional study. Result The plasma LRG1 level differed significantly according to the quartiles of urinary albumin excretion (Q1 [<7.7 mg/g], 17.1 μg/mL; Q2 [7.7-15.0 mg/g], 17.5 μg/mL; Q3 [15.1-61.4 mg/g], 18.6 μg/mL; Q4 [≥61.5 mg/g], 22.3 μg/mL; p for trend = 0.003) under adjustment with other covariates. A positive correlation was found between plasma LRG1 level and urinary albumin excretion (ρ = 0.256, p <0.001). According to a multivariate model, the association between LRG1 and urinary albumin excretion remained significant, under adjustment for confounding factors (β = 0.285, p <0.001). Conclusion Plasma LRG1 level was independently associated with urinary albumin excretion in patients with type 2 diabetes. This study suggests that LRG1 may be associated with increased excretion of urinary albumin in the early stages of diabetic nephropathy.
Collapse
Affiliation(s)
- Jin Ook Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Seon-Young Park
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Dong Hyeok Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Dong Jin Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Min Young Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
20
|
Liang J, Liu Y. Animal Models of Kidney Disease: Challenges and Perspectives. KIDNEY360 2023; 4:1479-1493. [PMID: 37526653 PMCID: PMC10617803 DOI: 10.34067/kid.0000000000000227] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Kidney disease is highly prevalent and affects approximately 850 million people worldwide. It is also associated with high morbidity and mortality, and current therapies are incurable and often ineffective. Animal models are indispensable for understanding the pathophysiology of various kidney diseases and for preclinically testing novel remedies. In the last two decades, rodents continue to be the most used models for imitating human kidney diseases, largely because of the increasing availability of many unique genetically modified mice. Despite many limitations and pitfalls, animal models play an essential and irreplaceable role in gaining novel insights into the mechanisms, pathologies, and therapeutic targets of kidney disease. In this review, we highlight commonly used animal models of kidney diseases by focusing on experimental AKI, CKD, and diabetic kidney disease. We briefly summarize the pathological characteristics, advantages, and drawbacks of some widely used models. Emerging animal models such as mini pig, salamander, zebrafish, and drosophila, as well as human-derived kidney organoids and kidney-on-a-chip are also discussed. Undoubtedly, careful selection and utilization of appropriate animal models is of vital importance in deciphering the mechanisms underlying nephropathies and evaluating the efficacy of new treatment options. Such studies will provide a solid foundation for future diagnosis, prevention, and treatment of human kidney diseases.
Collapse
Affiliation(s)
- Jianqing Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
21
|
Yu Q, Chen Y, Zhao Y, Huang S, Xin X, Jiang L, Wang H, Wu W, Qu L, Xiang C, Wang S, Liu G, Yang L. Nephropathy Is Aggravated by Fatty Acids in Diabetic Kidney Disease through Tubular Epithelial Cell Necroptosis and Is Alleviated by an RIPK-1 Inhibitor. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:408-423. [PMID: 37927402 PMCID: PMC10624943 DOI: 10.1159/000529995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 11/07/2023]
Abstract
Introduction Diabetic kidney disease (DKD), one of the leading causes of end-stage renal disease, has complex pathogenic mechanisms and few effective clinical therapies. DKD progression is accompanied by the loss of renal resident cells, followed by chronic inflammation and extracellular matrix deposition. Necroptosis is a newly discovered form of regulated cell death and is a major form of intrinsic cell loss in certain diabetic complications such as cardiomyopathy, intestinal disease, and retinal neuropathy; however, its significance in DKD is largely unknown. Methods In this study, the expression of necroptosis marker phosphorylated MLKL (p-MLKL) in renal biopsy tissues of patients with DKD was detected using immunofluorescence and semiquantified using immunohistochemistry. The effects of different disease-causing factors on necroptosis activation in human HK-2 cells were evaluated using immunofluorescence and Western blotting. db/db diabetic mice were fed a high-fat diet to establish an animal model of DKD with significant renal tubule damage. Mice were treated with the RIPK1 inhibitor RIPA-56 to evaluate its renal protective effects. mRNA transcriptome sequencing was used to explore the changes in signaling pathways after RIPA-56 treatment. Oil red O staining and electron macroscopy were used to observe lipid droplet accumulation in renal biopsy tissues and mouse kidney tissues. Results Immunostaining of phosphorylated RIPK1/RIPK3/MLKL verified the occurrence of necroptosis in renal tubular epithelial cells of patients with DKD. The level of the necroptosis marker p-MLKL correlated positively with the severity of renal functional, pathological damages, and lipid droplet accumulation in patients with DKD. High glucose and fatty acids were the main factors causing necroptosis in human renal tubular HK-2 cells. Renal function deterioration and renal pathological injury were accelerated, and the necroptosis pathway was activated in db/db mice fed a high-fat diet. Application of RIPA-56 effectively reduced the degree of renal injury, inhibited the necroptosis pathway activation, and reduced necroinflammation and lipid droplet accumulation in the renal tissues of db/db mice fed a high-fat diet. Conclusion The present study revealed the role of necroptosis in the progression of DKD and might provide a new therapeutic target for the treatment of DKD.
Collapse
Affiliation(s)
- Qi Yu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Ying Chen
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Youlu Zhao
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuo Huang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Xiaohong Xin
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Lei Jiang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Wenyan Wu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Lei Qu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Chengang Xiang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Gang Liu
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Renal Division, Renal Pathology Center, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of CKD Prevention and Treatment (Peking University), Ministry of Education of China, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Talukdar A, Basumatary M. Rodent models to study type 1 and type 2 diabetes induced human diabetic nephropathy. Mol Biol Rep 2023; 50:7759-7782. [PMID: 37458869 DOI: 10.1007/s11033-023-08621-z] [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: 04/26/2023] [Accepted: 06/21/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Diabetic nephropathy (DN), an outcome of prolonged diabetes, has affected millions of people worldwide and every year the incidence and prevalence increase substantially. The symptoms may start with mild manifestations of the disease such as increased albuminuria, serum creatinine levels, thickening of glomerular basement membrane, expansion of mesangial matrix to severe pathological symptoms such as glomerular lesions and tubulointerstitial fibrosis which may further proceed to cardiovascular dysfunction or end-stage renal disease. PERSPECTIVE Numerous therapeutic interventions are being explored for the management of DN, however, these interventions do not completely halt the progression of this disease and hence animal models are being explored to identify critical genetic and molecular parameters which could help in tackling the disease. Rodent models which mostly include mice and rats are commonly used experimental animals which provide a wide range of advantages in understanding the onset and progression of disease in humans and also their response to a wide range of interventions helps in the development of effective therapeutics. Rodent models of type 1 and type 2 diabetes induced DN have been developed utilizing different platforms and interventions during the last few decades some of which mimic various stages of diabetes ranging from early to later stages. However, a rodent model which replicates all the features of human DN is still lacking. This review tries to evaluate the rodent models that are currently available and understand their features and limitations which may help in further development of more robust models of human DN. CONCLUSION Using these rodent models can help to understand different aspects of human DN although further research is required to develop more robust models utilizing diverse genetic platforms which may, in turn, assist in developing effective interventions to target the disease at different levels.
Collapse
Affiliation(s)
- Amit Talukdar
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India.
| | - Mandira Basumatary
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India
| |
Collapse
|
23
|
Daehn IS, Ekperikpe US, Stadler K. Redox regulation in diabetic kidney disease. Am J Physiol Renal Physiol 2023; 325:F135-F149. [PMID: 37262088 PMCID: PMC10393330 DOI: 10.1152/ajprenal.00047.2023] [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: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/20/2023] [Indexed: 06/03/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the most devastating complications of diabetes mellitus, where currently there is no cure available. Several important mechanisms contribute to the pathogenesis of this complication, with oxidative stress being one of the key factors. The past decades have seen a large number of publications with various aspects of this topic; however, the specific details of redox regulation in DKD are still unclear. This is partly because redox biology is very complex, coupled with a complex and heterogeneous organ with numerous cell types. Furthermore, often times terms such as "oxidative stress" or reactive oxygen species are used as a general term to cover a wide and rich variety of reactive species and their differing reactions. However, no reactive species are the same, and not all of them are capable of biologically relevant reactions or "redox signaling." The goal of this review is to provide a biochemical background for an array of specific reactive oxygen species types with varying reactivity and specificity in the kidney as well as highlight some of the advances in redox biology that are paving the way to a better understanding of DKD development and risk.
Collapse
Affiliation(s)
- Ilse S Daehn
- Division of Nephrology, Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Ubong S Ekperikpe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| |
Collapse
|
24
|
Schürfeld R, Sandner B, Hoffmann A, Klöting N, Baratashvili E, Nowicki M, Paeschke S, Kosacka J, Kralisch S, Bachmann A, Frille A, Dietel A, Stolzenburg JU, Blüher M, Zhang MZ, Harris RC, Isermann B, Stumvoll M, Tönjes A, Ebert T. Renal function is a major predictor of circulating acyl-CoA-binding protein/diazepam-binding inhibitor. Front Endocrinol (Lausanne) 2023; 14:1152444. [PMID: 37288304 PMCID: PMC10242139 DOI: 10.3389/fendo.2023.1152444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Objective Acyl-CoA-binding protein (ACBP)/diazepam-binding inhibitor has lately been described as an endocrine factor affecting food intake and lipid metabolism. ACBP is dysregulated in catabolic/malnutrition states like sepsis or systemic inflammation. However, regulation of ACBP has not been investigated in conditions with impaired kidney function, so far. Design/methods Serum ACBP concentrations were investigated by enzyme-linked immunosorbent assay i) in a cohort of 60 individuals with kidney failure (KF) on chronic haemodialysis and compared to 60 individuals with a preserved kidney function; and ii) in a human model of acute kidney dysfunction (AKD). In addition, mACBP mRNA expression was assessed in two CKD mouse models and in two distinct groups of non-CKD mice. Further, mRNA expression of mACBP was measured in vitro in isolated, differentiated mouse adipocytes - brown and white - after exposure to the uremic agent indoxyl sulfate. Results Median [interquartile range] serum ACBP was almost 20-fold increased in KF (514.0 [339.3] µg/l) compared to subjects without KF (26.1 [39.1] µg/l) (p<0.001). eGFR was the most important, inverse predictor of circulating ACBP in multivariate analysis (standardized β=-0.839; p<0.001). Furthermore, AKD increased ACBP concentrations almost 3-fold (p<0.001). Increased ACBP levels were not caused by augmented mACBP mRNA expression in different tissues of CKD mice in vivo or in indoxyl sulfate-treated adipocytes in vitro. Conclusions Circulating ACBP inversely associates with renal function, most likely through renal retention of the cytokine. Future studies need to investigate ACBP physiology in malnutrition-related disease states, such as CKD, and to adjust for markers of renal function.
Collapse
Affiliation(s)
- Robin Schürfeld
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Benjamin Sandner
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Annett Hoffmann
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig and University Hospital, Leipzig, Germany
| | - Ekaterine Baratashvili
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Department of Cardiology, Angiology and Internal Intensive-Care Medicine, Klinikum St. Georg, Leipzig, Germany
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Sabine Paeschke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Joanna Kosacka
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Susan Kralisch
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Anette Bachmann
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Armin Frille
- Department of Respiratory Medicine, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Anja Dietel
- Department of Urology, University of Leipzig, Leipzig, Germany
| | | | - Matthias Blüher
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig and University Hospital, Leipzig, Germany
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Medicine, Nashville Veterans Affairs Hospital, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Raymond C. Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Medicine, Nashville Veterans Affairs Hospital, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Thomas Ebert
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
25
|
Juin SK, Ouseph R, Gondim DD, Jala VR, Sen U. Diabetic Nephropathy and Gaseous Modulators. Antioxidants (Basel) 2023; 12:antiox12051088. [PMID: 37237955 DOI: 10.3390/antiox12051088] [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: 03/07/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic nephropathy (DN) remains the leading cause of vascular morbidity and mortality in diabetes patients. Despite the progress in understanding the diabetic disease process and advanced management of nephropathy, a number of patients still progress to end-stage renal disease (ESRD). The underlying mechanism still needs to be clarified. Gaseous signaling molecules, so-called gasotransmitters, such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), have been shown to play an essential role in the development, progression, and ramification of DN depending on their availability and physiological actions. Although the studies on gasotransmitter regulations of DN are still emerging, the evidence revealed an aberrant level of gasotransmitters in patients with diabetes. In studies, different gasotransmitter donors have been implicated in ameliorating diabetic renal dysfunction. In this perspective, we summarized an overview of the recent advances in the physiological relevance of the gaseous molecules and their multifaceted interaction with other potential factors, such as extracellular matrix (ECM), in the severity modulation of DN. Moreover, the perspective of the present review highlights the possible therapeutic interventions of gasotransmitters in ameliorating this dreaded disease.
Collapse
Affiliation(s)
- Subir Kumar Juin
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Venkatakrishna Rao Jala
- Department of Microbiology & Immunology, Brown Cancer Center, Center for Microbiomics, Inflammation and Pathogenicity, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
26
|
Luo W, Tang S, Xiao X, Luo S, Yang Z, Huang W, Tang S. Translation Animal Models of Diabetic Kidney Disease: Biochemical and Histological Phenotypes, Advantages and Limitations. Diabetes Metab Syndr Obes 2023; 16:1297-1321. [PMID: 37179788 PMCID: PMC10168199 DOI: 10.2147/dmso.s408170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Animal models play a crucial role in studying the pathogenesis of diseases, developing new drugs, identifying disease risk markers, and improving means of prevention and treatment. However, modeling diabetic kidney disease (DKD) has posed a challenge for scientists. Although numerous models have been successfully developed, none of them can encompass all the key characteristics of human DKD. It is essential to choose the appropriate model according to the research needs, as different models develop different phenotypes and have their limitations. This paper provides a comprehensive overview of biochemical and histological phenotypes, modeling mechanisms, advantages and limitations of DKD animal models, in order to update relevant model information and provide insights and references for generating or selecting the appropriate animal models to fit different experimental needs.
Collapse
Affiliation(s)
- Wenting Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Xiang Xiao
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Simin Luo
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Zixuan Yang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| | - Wei Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Songqi Tang
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan Province, People’s Republic of China
| |
Collapse
|
27
|
Gao Y, Su X, Xue T, Zhang N. The beneficial effects of astragaloside IV on ameliorating diabetic kidney disease. Biomed Pharmacother 2023; 163:114598. [PMID: 37150034 DOI: 10.1016/j.biopha.2023.114598] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/22/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic kidney disease (DKD) has become the major cause of chronic kidney disease or end-stage renal disease. There is still a need for innovative treatment strategies for preventing, arresting, treating, and reversing DKD, and a plethora of scientific evidence has revealed that Chinese herbal monomers can attenuate DKD in multiple ways. Astragaloside IV (AS-IV) is one of the active ingredients of Astragalus membranaceus and was selected as a chemical marker in the Chinese Pharmacopeia for quality control purposes. An increasing amount of studies indicate that AS-IV is a promising novel drug for the treatment of DKD. AS-IV has been shown to improve DKD by combating oxidative stress, attenuating endoplasmic reticulum stress, regulating calcium homeostasis, alleviating inflammation, improving vascular function, improving epithelial to mesenchymal transition and so on. This review briefly summarizes the pathogenesis of DKD, systematically reviews the mechanisms by which AS-IV improves DKD, and aims to facilitate related pharmacological research and development to promote the utilization of Chinese herbal monomers in DKD.
Collapse
Affiliation(s)
- Yiwei Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Xin Su
- Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Taiqi Xue
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China.
| |
Collapse
|
28
|
Tao Y, Yazdizadeh Shotorbani P, Inman D, Das-Earl P, Ma R. Store-operated Ca 2+ entry inhibition ameliorates high glucose and ANG II-induced podocyte apoptosis and mitochondrial damage. Am J Physiol Renal Physiol 2023; 324:F494-F504. [PMID: 36995925 PMCID: PMC10151057 DOI: 10.1152/ajprenal.00297.2022] [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: 12/07/2022] [Revised: 03/10/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Hyperglycemia and increased activity of the renal angiotensin II (ANG II) system are two primary pathogenic stimuli for the onset and progression of podocyte injury in diabetic nephropathy. However, the underlying mechanisms are not fully understood. Store-operated Ca2+ entry (SOCE) is an important mechanism that helps maintain cell Ca2+ homeostasis in both excitable and nonexcitable cells. Our previous study demonstrated that high glucose (HG) enhanced podocyte SOCE (1). It is also known that ANG II activates SOCE by releasing endoplasmic reticulum Ca2+. However, the role of SOCE in stress-induced podocyte apoptosis and mitochondrial dysfunction remains unclear. The present study was aimed to determine whether enhanced SOCE mediated HG- and ANG II-induced podocyte apoptosis and mitochondrial damage. In kidneys of mice with diabetic nephropathy, the number of podocytes was significantly reduced. In cultured human podocytes, both HG and ANG II treatment induced podocyte apoptosis, which was significantly blunted by an SOCE inhibitor, BTP2. Seahorse analysis showed that podocyte oxidative phosphorylation in response to HG and ANG II was impaired. This impairment was significantly alleviated by BTP2. The SOCE inhibitor, but not a transient receptor potential cation channel subfamily C member 6 inhibitor, significantly blunted the damage of podocyte mitochondrial respiration induced by ANG II treatment. Furthermore, BTP2 reversed impaired mitochondrial membrane potential and ATP production and enhanced mitochondrial superoxide generation induced by HG treatment. Finally, BTP2 prevented the overwhelming Ca2+ uptake in HG-treated podocytes. Taken together, our results suggest that enhanced SOCE mediated HG- and ANG II-induced podocyte apoptosis and mitochondrial injury.NEW & NOTEWORTHY This study tested the hypothesis that overwhelming store-operated Ca2+ entry is a novel mechanism contributing to high glucose- and angiotensin II-induced podocyte apoptosis and mitochondrial injury.
Collapse
Affiliation(s)
- Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Parisa Yazdizadeh Shotorbani
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Denise Inman
- The North Texas Eye Research Institute and Department of Pharmaceutical Science, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Paromita Das-Earl
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
29
|
Tao Y, Young‐Stubbs C, Yazdizadeh Shotorbani P, Su D, Mathis KW, Ma R. Sex and strain differences in renal hemodynamics in mice. Physiol Rep 2023; 11:e15644. [PMID: 36946063 PMCID: PMC10031302 DOI: 10.14814/phy2.15644] [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: 02/13/2023] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 03/23/2023] Open
Abstract
The present study was to examine sex and strain differences in glomerular filtration rate (GFR) and renal blood flow (RBF) in C57BL6, 129/Sv, and C57BLKS/J mice, three commonly used mouse strains in renal research. GFR was measured by transdermal measurement of FITC-sinitrin clearance in conscious mice. RBF was measured by a flow probe placed in the renal artery under an anesthetic state. In C57BL6 mice, there were no sex differences in both GFR and RBF. In 129/Sv mice, females had significantly greater GFR than males at age of 24 weeks, but not at 8 weeks. However, males had higher RBF and lower renal vascular resistance (RVR). Similar to 129/Sv, female C57BLKS/J had significantly greater GFR at both 8 and 24 weeks, lower RBF, and higher RVR than males. Across strains, male 129/Sv had lower GFR and higher RBF than male C57BL6, but no significant difference in GFR and greater RBF than male C57BLKS/J. No significant difference in GFR or RBF was observed between C57BL6 and C57BLKS/J mice. Deletion of eNOS in C57BLKS/J mice reduced GFR in both sexes, but decreased RBF in males. Furthermore, there were no sex differences in the severity of renal injury in eNOS-/- dbdb mice. Taken together, our study suggests that sex differences in renal hemodynamics in mice are strain and age dependent. eNOS was not involved in the sex differences in GFR, but in RBF. Furthermore, the sexual dimorphism did not impact the severity of renal injury in diabetic nephropathy.
Collapse
Affiliation(s)
- Yu Tao
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Cassandra Young‐Stubbs
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | | | - Dong‐Ming Su
- Department of Microbiology, Immunology and GeneticsUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Keisa W. Mathis
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Rong Ma
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
30
|
Liu JJ, Liu S, Wang J, Pek SL, Lee J, Gurung RL, Ang K, Shao YM, Tavintharan S, Tang WE, Sum CF, Lim SC. Urine Leucine-Rich α-2 Glycoprotein 1 (LRG1) Predicts the Risk of Progression to End-Stage Kidney Disease in Patients With Type 2 Diabetes. Diabetes Care 2023; 46:408-415. [PMID: 36516193 PMCID: PMC9887617 DOI: 10.2337/dc22-1611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/09/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Leucine-rich α-2 glycoprotein 1 (LRG1) was recently identified as an amplifier of transforming growth factor-β (TGF-β)-induced kidney fibrosis in animal models. We aimed to study whether urine LRG1 is associated with risk of progression to end-stage kidney disease (ESKD) in individuals with type 2 diabetes. RESEARCH DESIGN AND METHODS A total of 1,837 participants with type 2 diabetes and estimated glomerular filtration rate (eGFR) >30 mL/min/1.73 m2 were recruited from a regional hospital and a primary care facility. Association of urine LRG1 with risk of ESKD (progression to sustained eGFR <15 mL/min/1.73 m2, dialysis, or death resulting from renal causes) was assessed by survival analyses. RESULTS During a median follow-up of 8.6 (interquartile range 5.8-9.6) years, 134 incident ESKD events were identified. Compared with those in the lowest tertile, participants with baseline urine LRG1 in the highest tertile had a 1.91-fold (95% CI 1.04-3.50) increased risk of progression to ESKD, after adjustment for cardiorenal risk factors, including eGFR and albuminuria. As a continuous variable, 1 SD increment in urine LRG1 was associated with a 1.53-fold (95% CI 1.19-1.98) adjusted risk of ESKD. Of note, the association of urine LRG1 with ESKD was independent of plasma LRG1. Moreover, urine LRG1 was associated with rapid kidney function decline and progression to macroalbuminuria, two common pathways leading to ESKD. CONCLUSIONS Urine LRG1, a TGF-β signaling modulator, predicts risk of progression to ESKD independently of clinical risk factors in patients with type 2 diabetes, suggesting that it may be a novel factor involved in the pathophysiological pathway leading to kidney disease progression.
Collapse
Affiliation(s)
- Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Sylvia Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Jiexun Wang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Janus Lee
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Keven Ang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Yi Ming Shao
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | | | - Chee Fang Sum
- Diabetes Center, Admiralty Medical Center, Singapore
| | - Su Chi Lim
- Diabetes Center, Admiralty Medical Center, Singapore
- Saw Swee Hock School of Public Heath, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Corresponding author: Su Chi Lim,
| |
Collapse
|
31
|
Saalbach A, Anderegg U, Wendt R, Beige J, Bachmann A, Klöting N, Blüher M, Zhang MZ, Harris RC, Stumvoll M, Tönjes A, Ebert T. Antifibrotic Soluble Thy-1 Correlates with Renal Dysfunction in Chronic Kidney Disease. Int J Mol Sci 2023; 24:1896. [PMID: 36768219 PMCID: PMC9916214 DOI: 10.3390/ijms24031896] [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: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Kidney fibrosis is a major culprit in the development and progression of chronic kidney disease (CKD), ultimately leading to the irreversible loss of organ function. Thymocyte differentiation antigen-1 (Thy-1) controls many core functions of fibroblasts relevant to fibrogenesis but is also found in a soluble form (sThy-1) in serum and urine. We investigated the association of sThy-1 with clinical parameters in patients with CKD receiving hemodialysis treatment compared to individuals with a preserved renal function. Furthermore, Thy-1 tissue expression was detected in a mouse model of diabetic CKD (eNOS-/-; db/db) and non-diabetic control mice (eNOS-/-). Serum and urinary sThy-1 concentrations significantly increased with deteriorating renal function, independent of the presence of diabetes. Serum creatinine is the major, independent, and inverse predictor of serum sThy-1 levels. Moreover, sThy-1 is not only predicted by markers of renal function but is also itself an independent and strong predictor of markers of renal function, i.e., serum creatinine. Mice with severe diabetic CKD show increased Thy-1 mRNA and protein expression in the kidney compared to control animals, as well as elevated urinary sThy-1 levels. Pro-fibrotic mediators, such as interleukin (IL)-4, IL-13, IL-6 and transforming growth factor β, increase Thy-1 gene expression and release of sThy-1 from fibroblasts. Our data underline the role of Thy-1 in the control of kidney fibrosis in CKD and raise the opportunity that Thy-1 may function as a renal antifibrotic factor.
Collapse
Affiliation(s)
- Anja Saalbach
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Ralph Wendt
- Hospital St. Georg, Division of Nephrology and Kuratorium for Dialysis and Transplantation, 04129 Leipzig, Germany
| | - Joachim Beige
- Hospital St. Georg, Division of Nephrology and Kuratorium for Dialysis and Transplantation, 04129 Leipzig, Germany
- Department for Internal Medicine, Medical Clinic 2, Martin-Luther-University Halle/Wittenberg, 06108 Halle, Germany
| | - Anette Bachmann
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
- Department of Medicine, Nashville Veterans Affairs Hospital, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | - Raymond C. Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
- Department of Medicine, Nashville Veterans Affairs Hospital, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | - Michael Stumvoll
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Anke Tönjes
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Thomas Ebert
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| |
Collapse
|
32
|
Jensen DM, Han P, Mangala LS, Lopez-Berestein G, Sood AK, Liu J, Kriegel AJ, Usa K, Widlansky ME, Liang M. Broad-acting therapeutic effects of miR-29b-chitosan on hypertension and diabetic complications. Mol Ther 2022; 30:3462-3476. [PMID: 35965413 PMCID: PMC9637778 DOI: 10.1016/j.ymthe.2022.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/18/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
MicroRNA miR-29 promotes endothelial function in human arterioles in part by targeting LYPLA1 and increasing nitric oxide production. In addition, miR-29 is a master inhibitor of extracellular matrix gene expression, which may attenuate fibrosis but could also weaken tissue structure. The goal of this study was to test whether miR-29 could be developed as an effective, broad-acting, and safe therapeutic. Substantial accumulation of miR-29b and effective knockdown of Lypla1 in several mouse tissues were achieved using a chitosan-packaged, chemically modified miR-29b mimic (miR-29b-CH-NP) injected systemically at 200 μg/kg body weight. miR-29b-CH-NP, injected once every 3 days, significantly attenuated angiotensin II-induced hypertension. In db/db mice, miR-29b-CH-NP treatment for 12 weeks decreased cardiac and renal fibrosis and urinary albuminuria. In uninephrectomized db/db mice, miR-29b-CH-NP treatment for 20 weeks significantly improved myocardial performance index and attenuated proteinuria. miR-29b-CH-NP did not worsen abdominal aortic aneurysm in ApoE knockout mice treated with angiotensin II. miR-29b-CH-NP caused aortic root fibrotic cap thinning in ApoE knockout mice fed a high-cholesterol and high-fat diet but did not worsen the necrotic zone or mortality. In conclusion, systemic delivery of low-dose miR-29b-CH-NP is an effective therapeutic for several forms of cardiovascular and renal disease in mice.
Collapse
Affiliation(s)
- David M Jensen
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Peng Han
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Liu
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alison J Kriegel
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kristie Usa
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael E Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mingyu Liang
- Department of Physiology, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
33
|
Harris RC. The Role of the Epidermal Growth Factor Receptor in Diabetic Kidney Disease. Cells 2022; 11:3416. [PMID: 36359813 PMCID: PMC9656309 DOI: 10.3390/cells11213416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 08/02/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is expressed in numerous cell types in the adult mammalian kidney and is activated by a family of EGF-like ligands. EGFR activation has been implicated in a variety of physiologic and pathophysiologic functions. There is increasing evidence that aberrant EGFR activation is a mediator of progressive kidney injury in diabetic kidney disease. This review will highlight recent studies indicating its potential role and mechanisms of injury of both glomerular and tubular cells in development and progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Raymond C. Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN 37232, USA; ; Tel.: +1-615-202-9426
- Tennessee and Veterans Affairs, Nashville, TN 37232, USA
| |
Collapse
|
34
|
Mechanism of Cornus Officinalis in Treating Diabetic Kidney Disease Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1799106. [PMID: 35855831 PMCID: PMC9288281 DOI: 10.1155/2022/1799106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 11/28/2022]
Abstract
Diabetic kidney disease (DKD), one of the most important diabetic complications, is a great clinical challenge. It still lacks proper therapeutic strategies without side effects due to the complex pathological mechanisms. Cornus officinalis (CO) is a common traditional Chinese medicine, which has been used in the treatment of DKD and takes beneficial effects in therapy. However, the mechanism of CO in treating DKD is not clear yet. In this study, network pharmacology was applied to illustrate the potential mechanism of CO and the interaction between targets of CO and targets of disease. First, the active ingredients of CO and related targets were screened from the online database. Second, the intersection network between CO and disease was constructed, and protein–protein interaction analysis was done. Third, GO and KEGG analysis were employed to figure out the key targets of CO. Finally, molecular docking was carried out in the software SYBYL to verify the effectiveness of the ingredients and targets selected. According to GO and KEGG analysis, drug metabolism-cytochrome P450, sphingolipid signaling pathway, HIF-1 signaling pathway, TGF-beta signaling pathway, cGMP-PKG signaling pathway, estrogen signaling pathway, and TNF signaling pathway were most closely related to the pathogenesis of DKD. Moreover, NOS3, TNF, ROCK1, PPARG, KDR, and HIF1A were identified as key targets in regulating the occurrence and development of the disease. This study provides evidence to elucidate the mechanism of CO comprehensively and systematically and lays the foundation for further research on CO.
Collapse
|
35
|
Bian Y, Shi C, Song S, Mu L, Wu M, Qiu D, Dong J, Zhang W, Yuan C, Wang D, Zhou Z, Dong X, Shi Y. Sestrin2 attenuates renal damage by regulating Hippo pathway in diabetic nephropathy. Cell Tissue Res 2022; 390:93-112. [PMID: 35821438 DOI: 10.1007/s00441-022-03668-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/01/2022] [Indexed: 11/27/2022]
Abstract
Glomerular mesangial cell proliferation and extracellular matrix accumulation contribute to the progression of diabetic nephropathy (DN). As a conserved stress-inducible protein, sestrin2 (Sesn2) plays critical role in the regulation of oxidative stress, inflammation, autophagy, metabolism, and endoplasmic reticulum stress. In this study, we investigated the role of Sesn2 on renal damage in diabetic kidney using transgenic mice overexpressing Sesn2 and the effect of Sesn2 on mesangial cell proliferation and extracellular matrix accumulation in diabetic conditions and the possible molecular mechanisms involved. Sesn2 overexpression improved renal function and decreased glomerular hypertrophy, albuminuria, mesangial expansion, extracellular matrix accumulation, and TGF-β1 expression, as well as oxidative stress in diabetic mice. In vitro experiments, using human mesangial cells (HMCs), revealed that Sesn2 overexpression inhibited high glucose (HG)-induced proliferation, fibronectin and collagen IV production, and ROS generation. Meanwhile, Sesn2 overexpression restored phosphorylation levels of Lats1 and YAP and inhibited TEAD1 expression. Inhibition of Lats1 accelerated HG-induced proliferation and expression of fibronectin and collagen IV. Verteporfin, an inhibitor of YAP, suppressed HG-induced proliferation and expression of fibronectin and collagen IV. However, Sesn2 overexpression reversed Lats1 deficiency-induced Lats1 and YAP phosphorylation, nuclear expression levels of YAP and TEAD1, and proliferation and fibronectin and collagen IV expressions in HMCs exposed to HG. In addition, antioxidant NAC or tempol treatment promoted phosphorylation of Lats1 and YAP and inhibited TEAD1 expression, proliferation, and fibronectin and collagen IV accumulation in HG-treated HMCs. Taken together, Sesn2 overexpression inhibited mesangial cell proliferation and fibrosis via regulating Hippo pathway in diabetic nephropathy. Induction of Sesn2 may be a potential therapeutic target in diabetic nephropathy.
Collapse
Affiliation(s)
- Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Chonglin Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lin Mu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Duojun Qiu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiajia Dong
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Wei Zhang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Chen Yuan
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Dongyun Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zihui Zhou
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuan Dong
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China.
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
36
|
Chaudhari S, Yazdizadeh Shotorbani P, Tao Y, Kasetti R, Zode G, Mathis KW, Ma R. Neogenin pathway positively regulates fibronectin production by glomerular mesangial cells. Am J Physiol Cell Physiol 2022; 323:C226-C235. [PMID: 35704698 DOI: 10.1152/ajpcell.00359.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neogenin, a transmembrane receptor, was recently found in kidney cells and immune cells. However, the function of neogenin signaling in kidney is not clear. Mesangial cells (MCs) are a major source of extracellular matrix (ECM) proteins in glomerulus. In many kidney diseases, MCs are impaired and manifest myofibroblast phenotype. Over production of ECM by the injured MCs promotes renal injury and accelerates the progression of kidney diseases. The present study was aimed to determine if neogenin receptor was expressed in MCs and if the receptor signaling regulated ECM protein production by MCs. We showed that neogenin was expressed in the glomerular MCs. Deletion of neogenin using CRISPR/Cas9 lentivirus system, significantly reduced the abundance of fibronectin, an ECM protein. Netrin-1, a ligand for neogenin, also significantly decreased fibronectin production by MCs and decreased neogenin protein expression in MCs. Furthermore, treatment of human MCs with high glucose (25 mM) significantly increased the protein abundance of neogenin as early as 8 h. Consistently, neogenin expression in glomerulus significantly increased in the eNOS-/- db/db diabetic mice starting as early as the age of 8 weeks and this increase sustained at least to the age of 24 weeks. We further found that the HG induced increase in neogenin abundance was blunted by antioxidant PEG-catalase and N-acetyl cysteine. Taken together, our results suggest a new mechanism of regulation of fibronectin production by MCs. This previously unrecognized neogenin-fibronectin pathway may contribute to glomerular injury responses during the course of diabetic nephropathy.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | | | - Yu Tao
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Ramesh Kasetti
- The North Texas Eye Research Institute and Dept. of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, United States
| | - Gulab Zode
- The North Texas Eye Research Institute and Dept. of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, United States
| | - Keisa W Mathis
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Rong Ma
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
37
|
Vargas-George S, Dave KR. Models of cerebral amyloid angiopathy-related intracerebral hemorrhage. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
38
|
IGFBP5 promotes diabetic kidney disease progression by enhancing PFKFB3-mediated endothelial glycolysis. Cell Death Dis 2022; 13:340. [PMID: 35418167 PMCID: PMC9007962 DOI: 10.1038/s41419-022-04803-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
Abstract
Renal inflammation is a critical pathophysiological characteristic of diabetic kidney disease (DKD). The mechanism of the inflammatory response is complicated, and there are few effective treatments for renal inflammation that can be used clinically. Insulin-like growth factor-binding protein 5 (IGFBP5) is an important secretory protein that is related to inflammation and fibrosis in several tissues. Studies have shown that the IGFBP5 level is significantly upregulated in DKD. However, the function of IGFBP5 and its mechanism in DKD remain unclear. Here, we showed that IGFBP5 levels were significantly increased in the kidneys of diabetic mice. Ablation of IGFBP5 alleviated kidney inflammation in DKD mice. Mechanistically, IGFBP5 increased glycolysis, which was characterized by increases in lactic acid and the extracellular acidification rate, by activating the transcription factor early growth response 1 (EGR1) and enhancing the expression of PFKFB3 in endothelial cells. Furthermore, a mutation in PFKFB3 attenuated renal inflammation in DKD mice. Taken together, we provided evidence that IGFBP5 enhanced kidney inflammation through metabolic reprogramming of glomerular endothelial cells. Our results provide new mechanistic insights into the effect of IGFBP5 on kidney and highlight potential therapeutic opportunities for IGFBP5 and the metabolic regulators involved in DKD. ![]()
Collapse
|
39
|
Veron D, Aggarwal PK, Li Q, Moeckel G, Kashgarian M, Tufro A. Podocyte VEGF-A Knockdown Induces Diffuse Glomerulosclerosis in Diabetic and in eNOS Knockout Mice. Front Pharmacol 2022; 12:788886. [PMID: 35280251 PMCID: PMC8906751 DOI: 10.3389/fphar.2021.788886] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor-a (VEGF-A) and nitric oxide (NO) are essential for glomerular filtration barrier homeostasis, and are dysregulated in diabetic kidney disease (DKD). While NO availability is consistently low in diabetes, both high and low VEGF-A have been reported in patients with DKD. Here we examined the effect of inducible podocyte VEGF-A knockdown (VEGFKD) in diabetic mice and in endothelial nitric oxide synthase knockout mice (eNOS−/−). Diabetes was induced with streptozotocin using the Animal Models of Diabetic Complications Consortium (AMDCC) protocol. Induction of podocyte VEGFKD led to diffuse glomerulosclerosis, foot process effacement, and GBM thickening in both diabetic mice with intact eNOS and in non-diabetic eNOS−/−:VEGFKD mice. VEGFKD diabetic mice developed mild proteinuria and maintained normal glomerular filtration rate (GFR), associated with extremely high NO and thiol urinary excretion. In eNOS−/−:VEGFKD (+dox) mice severe diffuse glomerulosclerosis was associated with microaneurisms, arteriolar hyalinosis, massive proteinuria, and renal failure. Collectively, data indicate that combined podocyte VEGF-A and eNOS deficiency result in diffuse glomerulosclerosis in mice; compensatory NO and thiol generation prevents severe proteinuria and GFR loss in VEGFKD diabetic mice with intact eNOS, whereas VEGFKD induction in eNOS−/−:VEGFKD mice causes massive proteinuria and renal failure mimicking DKD in the absence of diabetes. Mechanistically, we identify VEGFKD-induced abnormal S-nitrosylation of specific proteins, including β3-integrin, laminin, and S-nitrosoglutathione reductase (GSNOR), as targetable molecular mechanisms involved in the development of advanced diffuse glomerulosclerosis and renal failure.
Collapse
Affiliation(s)
- Delma Veron
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Pardeep K Aggarwal
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Qi Li
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Alda Tufro
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
40
|
Shinozaki Y, Katayama Y, Yamaguchi F, Suzuki T, Watanabe K, Uno K, Tsutsui T, Sugimoto M, Shinohara M, Miyajima K, Ohta T. Salt loading with unilateral nephrectomy accelerates decline in glomerular filtration rate in the hypertensive, obese, type 2 diabetic SDT fatty rat model of diabetic kidney disease. Clin Exp Pharmacol Physiol 2022; 49:492-500. [DOI: 10.1111/1440-1681.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Yuichi Shinozaki
- Laboratory of Animal Physiology and Functional Anatomy Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Yuko Katayama
- Research Division SCOHIA PHARMA, Inc. Kanagawa Japan
| | | | | | - Kana Watanabe
- Department of Nutritional Science and Food Safety Faculty of Applied Biosciences Tokyo University of Agriculture Tokyo Japan
| | - Kinuko Uno
- Department of Nutritional Science and Food Safety Faculty of Applied Biosciences Tokyo University of Agriculture Tokyo Japan
| | - Takahiro Tsutsui
- Laboratory of Animal Physiology and Functional Anatomy Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Miki Sugimoto
- Laboratory of Animal Physiology and Functional Anatomy Graduate School of Agriculture Kyoto University Kyoto Japan
| | | | - Katsuhiro Miyajima
- Department of Nutritional Science and Food Safety Faculty of Applied Biosciences Tokyo University of Agriculture Tokyo Japan
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy Graduate School of Agriculture Kyoto University Kyoto Japan
| |
Collapse
|
41
|
Abstract
Diabetes mellitus (DM) is gradually attacking the health and life of people all over the world. Diabetic kidney disease (DKD) is one of the most common chronic microvascular complications of DM, whose mechanism is complex and still lacks research. Sirtuin family is a class III histone deacetylase with highly conserved NAD+ binding domain and catalytic functional domain, while different N-terminal and C-terminal structures enable them to bind different deacetylated substrates to participate in the cellular NAD+ metabolism. The kidney is an organ rich in NAD+ and database exploration of literature shows that the Sirtuin family has different expression localization in renal, cellular, and subcellular structures. With the progress of modern technology, a variety of animal models and reagents for the Sirtuin family and DKD emerged. Machine learning in the literature shows that the Sirtuin family can regulate pathophysiological injury mainly in the glomerular filtration membrane, renal tubular absorption, and immune inflammation through various mechanisms such as epigenetics, multiple signaling pathways, and mitochondrial function. These mechanisms are the key nodes participating in DKD. Thus, it is of great significance for target therapy to study biological functions of the Sirtuin family and DKD regulation mechanism in-depth.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- *Correspondence: Huiwen Ren,
| |
Collapse
|
42
|
Kotake H, Yamada S, Ogura Y, Watanabe S, Inoue K, Ichikawa D, Sugaya T, Ohata K, Natsuki Y, Hoshino S, Watanabe M, Kimura K, Shibagaki Y, Kamijo-Ikemori A. Endurance exercise training-attenuated diabetic kidney disease with muscle weakness in spontaneously diabetic Torii fatty rats. Kidney Blood Press Res 2021; 47:203-218. [PMID: 34923497 DOI: 10.1159/000521464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/11/2021] [Indexed: 11/19/2022] Open
Abstract
Background The aim of this study was to evaluate protective effects of endurance exercise training against diabetic kidney disease (DKD) with muscle weakness by using male spontaneously diabetic Torii (SDT) fatty rats as type 2 diabetic animal models with obesity, hypertension, and hyperlipidemia. Methods Eight-week-old SDT fatty rats (n = 12) and Sprague-Dawley (SD) rats (n = 10) were randomly divided into exercise (Ex; SDT-Ex: n = 6, SD-Ex: n = 5) and sedentary groups (SDT-Cont: n = 6, SD-Cont: n = 5), respectively. Each group underwent regular treadmill exercise four times a week from ages 8 to 16 weeks. Results The exercise attenuated hypertension and hyperlipidemia and prevented increases in renal parameter levels without affecting blood glucose levels. In the SDT fatty rats, it prevented induction of renal morphological abnormalities in the interstitium of the superficial and intermediate layers of the cortex. Downregulated expression of endothelial nitric oxide synthase in the glomerulus of the SDT fatty rats was significantly upregulated by the exercise. The exercise upregulated the renal expressions of both medium-chain acyl-CoA dehydrogenase and peroxisome proliferator-activated receptor γ coactivator-1α related to fatty acid metabolism. It increased muscle strength and both muscle weight and cross-sectional area of type IIb muscle fibers in the extensor digitorum longus muscle in the SDT fatty rats. Conclusion Endurance exercise training in type 2 diabetes ameliorates DKD by improving endothelial abnormality and enhancing fatty acid metabolism in addition to attenuated hypertension, hyperlipidemia, and muscle weakness independently of blood glucose levels.
Collapse
Affiliation(s)
- Hitoshi Kotake
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shohei Yamada
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shiika Watanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kazuho Inoue
- Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Keiichi Ohata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yasunori Natsuki
- Institute for Ultrastructural Morphology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Minoru Watanabe
- Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | | | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
- Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Japan
- Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| |
Collapse
|
43
|
Repurposing Riociguat to Target a Novel Paracrine Nitric Oxide-TRPC6 Pathway to Prevent Podocyte Injury. Int J Mol Sci 2021; 22:ijms222212485. [PMID: 34830371 PMCID: PMC8621407 DOI: 10.3390/ijms222212485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increased expression and activity of the Ca2+ channel transient receptor potential channel 6 (TRPC6) is associated with focal segmental glomerulosclerosis, but therapeutic strategies to target TRPC6 are currently lacking. Nitric oxide (NO) is crucial for normal glomerular function and plays a protective role in preventing glomerular diseases. We investigated if NO prevents podocyte injury by inhibiting injurious TRPC6-mediated signaling in a soluble guanylate cyclase (sGC)-dependent manner and studied the therapeutic potential of the sGC stimulator Riociguat. Experiments were performed using human glomerular endothelial cells and podocytes. Podocyte injury was induced by Adriamycin incubation for 24 h, with or without the NO-donor S-Nitroso-N-acetyl-DL-penicillamine (SNAP), the sGC stimulator Riociguat or the TRPC6 inhibitor Larixyl Acetate (LA). NO and Riociguat stimulated cGMP synthesis in podocytes, decreased Adriamycin-induced TRPC6 expression, inhibited the Adriamycin-induced TRPC6-mediated Ca2+ influx and reduced podocyte injury. The protective effects of Riociguat and NO were blocked when sGC activity was inhibited with 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or when TRPC6 activity was inhibited by LA. Our data demonstrate a glomerular (e)NOS-NO-sGC-cGMP-TRPC6 pathway that prevents podocyte injury, which can be translated to future clinical use by, e.g., repurposing the market-approved drug Riociguat.
Collapse
|
44
|
Gouliopoulos N, Siasos G, Oikonomou D, Oikonomou E, Konsola T, Kollia C, Athanasiou D, Dimitropoulos S, Rouvas A, Kassi E, Tousoulis D, Moschos MM. The association of T786C and G894T polymorphisms of eNOS gene with diabetic retinopathy in Greece. Eur J Ophthalmol 2021; 32:2582-2588. [PMID: 34779284 DOI: 10.1177/11206721211054724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To investigate whether eNOS T786C (rs2070744) and G894T (rs1799983) gene polymorphisms are associated with diabetic retinopathy in Greek diabetic patients. MATERIALS 271 patients with type-2 diabetes mellitus participated in our study; 130 suffered from diabetic retinopathy and 141 not. All the patients underwent a complete ophthalmological examination, while clinical and demographic data were assessed. Furthermore, they were genotyped for rs2070744 and rs1799983 single nucleotide polymorphisms of eNOS gene. RESULTS Regarding the clinical and demographic data, no significant differences were detected between the studied groups, except for hemoglobin A1c levels and the frequency of insulin treatment (higher in patients with diabetic retinopathy). The frequency of rs1799983 GT genotype was significantly elevated in patients with diabetic retinopathy (55% vs. 40%, P = 0.011) and was associated with a 2-fold increased risk of developing retinopathy (OR 1.92, 95% CI 1.16-3.17). Furthermore, we demonstrated that the aforementioned genotype was significantly and independently associated with increased odds for retinopathy onset in diabetic subjects (OR 2.23, 95% CI 1.28-3.90, P = 0.005), regardless of the impact of other confounders. CONCLUSIONS We documented that rs1799983 GT genotype could be recognized as an independent risk factor of retinopathy in Greek patients with type-2 diabetes mellitus, while no role for rs2070744 polymorphism was identified. Further research in different ethnic groups will clarify the exact association of these polymorphisms with the risk for diabetic retinopathy development.
Collapse
Affiliation(s)
- Nikolaos Gouliopoulos
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece.,1st Department of Ophthalmology, University of Athens Medical School, G. Gennimatas Hospital, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Dimitra Oikonomou
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Theodosia Konsola
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Christina Kollia
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Dimitris Athanasiou
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Stathis Dimitropoulos
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Alexandros Rouvas
- 2nd Department of Ophthalmology, University of Athens Medical School, 69038Attikon Hospital, Athens, Greece
| | - Eva Kassi
- 1st Department of Propaedeutic and Internal Medicine, Division of Diabetes, 68989University of Athens Medical School, Laiko Hospital, Athens, Greece
| | - Dimitrios Tousoulis
- 1st Department of Cardiology, 393206University of Athens Medical School, Hippokration Hospital, Athens, Greece
| | - Marilita M Moschos
- 1st Department of Ophthalmology, University of Athens Medical School, G. Gennimatas Hospital, Athens, Greece
| |
Collapse
|
45
|
Ventricular arrhythmias in mouse models of diabetic kidney disease. Sci Rep 2021; 11:20570. [PMID: 34663875 PMCID: PMC8523538 DOI: 10.1038/s41598-021-99891-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/05/2022] Open
Abstract
Chronic kidney disease (CKD) affects more than 20 million people in the US, and it is associated with a significantly increased risk of sudden cardiac death (SCD). Despite the significance, the mechanistic relationship between SCD and CKD is not clear and there are few effective therapies. Using optical mapping techniques, we tested the hypothesis that mouse models of progressive diabetic kidney disease (DKD) exhibit enhanced ventricular arrhythmia incidence and underlying arrhythmia substrates. Compared to wild-type mice, both Leprdb/db eNOS−/− (2KO) and high fat diet plus low dose streptozotocin (HFD + STZ) mouse models of DKD experienced sudden death and greater arrhythmia inducibility, which was more common with isoproterenol than programmed electrical stimulation. 2KO mice demonstrated slowed conduction velocity, prolonged action potential duration (APD), and myocardial fibrosis; both 2KO and HFD + STZ mice exhibited arrhythmias and calcium dysregulation with isoproterenol challenge. Finally, circulating concentrations of the uremic toxin asymmetric dimethylarginine (ADMA) were elevated in 2KO mice. Incubation of human cardiac myocytes with ADMA prolonged APD, as also observed in 2KO mice hearts ex vivo. The present study elucidates an arrhythmia-associated mechanism of sudden death associated with DKD, which may lead to more effective treatments in the vulnerable DKD patient population.
Collapse
|
46
|
Moreira LDSG, Fanton S, Cardozo L, Borges NA, Combet E, Shiels PG, Stenvinkel P, Mafra D. Pink pressure: beetroot (Beta vulgaris rubra) as a possible novel medical therapy for chronic kidney disease. Nutr Rev 2021; 80:1041-1061. [PMID: 34613396 DOI: 10.1093/nutrit/nuab074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) manifests with systemic inflammation, oxidative stress, and gut dysbiosis, resulting in metabolic disorders and elevated rates of cardiovascular disease-associated death. These all correlate with a high economic cost to healthcare systems. Growing evidence indicates that diet is an indispensable ally in the prevention and management of CKD and its complications. In this context, the root vegetable beetroot (Beta vulgaris rubra) deserves special attention because it is a source of several bioactive compounds, such as nitrate, betaine, and betalain, and has shown beneficial effects in CKD, including reduction of blood pressure, anti-inflammatory effects, and antioxidant actions by scavenging radical oxidative species, as observed in preclinical studies. Beetroot consumption as a possible therapeutic strategy to improve the clinical treatment of patients with CKD and future directions for clinical studies are addressed in this narrative review.
Collapse
Affiliation(s)
- Laís de Souza Gouveia Moreira
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Susane Fanton
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ludmila Cardozo
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Natalia A Borges
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Emilie Combet
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Paul G Shiels
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Stenvinkel
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Denise Mafra
- L.d.S.G. Moreira and D. Mafra are with the Graduate Program in Medical Sciences, Fluminense Federal University, Niterói, Rio de Janiero, Brazil. S. Fanton, L. Cardozo, and D. Mafra are with the Graduate Program in Cardiovascular Sciences, Federal Fluminense University, Niterói-Rio de Janeiro, RJ, Brazil. N.A. Borges is with the Institute of Nutrition, State University of Rio de Janeiro, Rio de Janeiro, Brazil. E. Combet is with the School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom. P.G. Shiels is with the Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom. P. Stenvinkel is with the Department of Renal Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
47
|
Wang Y, Niu A, Pan Y, Cao S, Terker AS, Wang S, Fan X, Toth CL, Ramirez Solano MA, Michell DL, Contreras D, Allen RM, Zhu W, Sheng Q, Fogo AB, Vickers KC, Zhang MZ, Harris RC. Profile of Podocyte Translatome During Development of Type 2 and Type 1 Diabetic Nephropathy Using Podocyte-Specific TRAP mRNA RNA-seq. Diabetes 2021; 70:2377-2390. [PMID: 34233930 PMCID: PMC8576501 DOI: 10.2337/db21-0110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/29/2021] [Indexed: 12/22/2022]
Abstract
Podocyte injury is important in development of diabetic nephropathy (DN). Although several studies have reported single-cell-based RNA sequencing (RNA-seq) of podocytes in type 1 DN (T1DN), the podocyte translating mRNA profile in type 2 DN (T2DN) has not previously been compared with that of T1DN. We analyzed the podocyte translatome in T2DN in podocin-Cre; Rosa26fsTRAP; eNOS-/-; db/db mice and compared it with that of streptozotocin-induced T1DN in podocin-Cre; Rosa26fsTRAP; eNOS-/- mice using translating ribosome affinity purification (TRAP) and RNA-seq. More than 125 genes were highly enriched in the podocyte ribosome. More podocyte TRAP genes were differentially expressed in T2DN than in T1DN. TGF-β signaling pathway genes were upregulated, while MAPK pathway genes were downregulated only in T2DN, while ATP binding and cAMP-mediated signaling genes were downregulated only in T1DN. Genes regulating actin filament organization and apoptosis increased, while genes regulating VEGFR signaling and glomerular basement membrane components decreased in both type 1 and type 2 diabetic podocytes. A number of diabetes-induced genes not previously linked to podocyte injury were confirmed in both mouse and human DN. On the basis of differences and similarities in the podocyte translatome in T2DN and T1DN, investigators can identify factors underlying the pathophysiology of DN and novel therapeutic targets to treat diabetes-induced podocyte injury.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Gene Expression Profiling
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Organ Specificity/genetics
- Podocytes/metabolism
- Podocytes/pathology
- Protein Biosynthesis/genetics
- Proteome/analysis
- Proteome/genetics
- Proteome/metabolism
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Seq
- Sequence Analysis, RNA
- Streptozocin
- Transcriptome
Collapse
Affiliation(s)
- Yinqiu Wang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Aolei Niu
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Yu Pan
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Shirong Cao
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Suwan Wang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Xiaofeng Fan
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Cynthia L Toth
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Marisol A Ramirez Solano
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Danielle L Michell
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Danielle Contreras
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Ryan M Allen
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Wanying Zhu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Quanhu Sheng
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Agnes B Fogo
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Kasey C Vickers
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, Nashville, TN
- Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Nashville, TN
| |
Collapse
|
48
|
Oe Y, Miyazaki M, Takahashi N. Coagulation, Protease-Activated Receptors, and Diabetic Kidney Disease: Lessons from eNOS-Deficient Mice. TOHOKU J EXP MED 2021; 255:1-8. [PMID: 34511578 DOI: 10.1620/tjem.255.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) dysfunction is known to exacerbate the progression and prognosis of diabetic kidney disease (DKD). One of the mechanisms through which this is achieved is that low eNOS levels are associated with hypercoagulability, which promotes kidney injury. In the extrinsic coagulation cascade, the tissue factor (factor III) and downstream coagulation factors, such as active factor X (FXa), exacerbate inflammation through activation of the protease-activated receptors (PARs). Recently, it has been shown that the lack of or reduced eNOS expression in diabetic mice, as a model of advanced DKD, increases renal tissue factor levels and PAR1 and 2 expression in their kidneys. Furthermore, pharmaceutical inhibition or genetic deletion of coagulation factors or PARs ameliorated inflammation in DKD in mice lacking eNOS. In this review, we summarize the relationship between eNOS, coagulation, and PARs and propose a novel therapeutic option for the management of patients with DKD.
Collapse
Affiliation(s)
- Yuji Oe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences
| |
Collapse
|
49
|
Eid SA, Hinder LM, Zhang H, Eksi R, Nair V, Eddy S, Eichinger F, Park M, Saha J, Berthier CC, Jagadish HV, Guan Y, Pennathur S, Hur J, Kretzler M, Feldman EL, Brosius FC. Gene expression profiles of diabetic kidney disease and neuropathy in eNOS knockout mice: Predictors of pathology and RAS blockade effects. FASEB J 2021; 35:e21467. [PMID: 33788970 DOI: 10.1096/fj.202002387r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/31/2022]
Abstract
Diabetic kidney disease (DKD) and diabetic peripheral neuropathy (DPN) are two common diabetic complications. However, their pathogenesis remains elusive and current therapies are only modestly effective. We evaluated genome-wide expression to identify pathways involved in DKD and DPN progression in db/db eNOS-/- mice receiving renin-angiotensin-aldosterone system (RAS)-blocking drugs to mimic the current standard of care for DKD patients. Diabetes and eNOS deletion worsened DKD, which improved with RAS treatment. Diabetes also induced DPN, which was not affected by eNOS deletion or RAS blockade. Given the multiple factors affecting DKD and the graded differences in disease severity across mouse groups, an automatic data analysis method, SOM, or self-organizing map was used to elucidate glomerular transcriptional changes associated with DKD, whereas pairwise bioinformatic analysis was used for DPN. These analyses revealed that enhanced gene expression in several pro-inflammatory networks and reduced expression of development genes correlated with worsening DKD. Although RAS treatment ameliorated the nephropathy phenotype, it did not alter the more abnormal gene expression changes in kidney. Moreover, RAS exacerbated expression of genes related to inflammation and oxidant generation in peripheral nerves. The graded increase in inflammatory gene expression and decrease in development gene expression with DKD progression underline the potentially important role of these pathways in DKD pathogenesis. Since RAS blockers worsened this gene expression pattern in both DKD and DPN, it may partly explain the inadequate therapeutic efficacy of such blockers.
Collapse
Affiliation(s)
- Stephanie A Eid
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lucy M Hinder
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hongyu Zhang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ridvan Eksi
- Department of Computational Medicine and Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Viji Nair
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sean Eddy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Felix Eichinger
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Meeyoung Park
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jharna Saha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Celine C Berthier
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hosagrahar V Jagadish
- Department of Computational Medicine and Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yuanfang Guan
- Department of Computational Medicine and Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Computational Medicine and Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Frank C Brosius
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
50
|
Daehn IS, Duffield JS. The glomerular filtration barrier: a structural target for novel kidney therapies. Nat Rev Drug Discov 2021; 20:770-788. [PMID: 34262140 PMCID: PMC8278373 DOI: 10.1038/s41573-021-00242-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 12/19/2022]
Abstract
Loss of normal kidney function affects more than 10% of the population and contributes to morbidity and mortality. Kidney diseases are currently treated with immunosuppressive agents, antihypertensives and diuretics with partial but limited success. Most kidney disease is characterized by breakdown of the glomerular filtration barrier (GFB). Specialized podocyte cells maintain the GFB, and structure-function experiments and studies of intercellular communication between the podocytes and other GFB cells, combined with advances from genetics and genomics, have laid the groundwork for a new generation of therapies that directly intervene at the GFB. These include inhibitors of apolipoprotein L1 (APOL1), short transient receptor potential channels (TRPCs), soluble fms-like tyrosine kinase 1 (sFLT1; also known as soluble vascular endothelial growth factor receptor 1), roundabout homologue 2 (ROBO2), endothelin receptor A, soluble urokinase plasminogen activator surface receptor (suPAR) and substrate intermediates for coenzyme Q10 (CoQ10). These molecular targets converge on two key components of GFB biology: mitochondrial function and the actin-myosin contractile machinery. This Review discusses therapies and developments focused on maintaining GFB integrity, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Ilse S Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jeremy S Duffield
- Research and Development, Prime Medicine, Cambridge, MA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|