1
|
Ren Z, Shao F, Chen S, Sun Y, Ding Z, Dong L, Zhang J, Zang Y. Contribution of alterations in peritubular capillary density and microcirculation to the progression of tubular injury and kidney fibrosis. J Pathol 2025; 266:95-108. [PMID: 40103536 DOI: 10.1002/path.6414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/18/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
Peritubular capillary (PTC) rarefaction is a common pathological feature of chronic kidney disease (CKD). The critical function of PTCs in maintaining blood supply for tubular epithelial cells renders PTCs a promising therapeutic target. However, the role of PTC rarefaction in the progression of kidney fibrosis remains elusive. In this study, we first characterized mice with altered PTC density. CD31 staining, together with microvascular network perfusion with FITC-labelled albumin and laser speckle contrast imaging, revealed a significant increase in PTC density in Flt1 heterozygous-deficient mice, whereas homozygous disruption of the plasminogen activator, urokinase receptor gene (Plaur/uPAR), led to a notable decrease in PTC density. Using these genetically distinct mice, we showed that preexisting higher PTC density protected against tubular injury and attenuated the progression of tubulointerstitial fibrosis in two distinct kidney injury models, namely, ischemia-reperfusion injury (IRI) and unilateral ureteral obstruction (UUO). By contrast, Plaur-deficient mice with established lower PTC density displayed exacerbated tubular injury and renal fibrosis when subjected to IRI or UUO. The pathophysiological significance of PTC density was associated with protective effects on tubular cell apoptosis and concomitant regeneration. Finally, vasodilation of the renal capillary with minoxidil, a clinically available drug, effectively prevented UUO-induced tubular injury and renal fibrosis. Moreover, minoxidil treatment abolished the detrimental effect of Plaur deficiency on the UUO-treated kidney, thus suggesting a causative role of PTC density in the susceptibility of Plaur knockout mice to tubular injury following fibrosis. Our results provide an overview of the pathologic significance of PTC density alterations in the progression of CKD, and show that improving peritubular microcirculation is effective in preventing tubular injury and the subsequent renal fibrosis. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Zhengrong Ren
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Fang Shao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Shuli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Yanyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| |
Collapse
|
2
|
Hironaka J, Okada H, Kusaba T, Nakajima H, Ushigome E, Hamaguchi M, Kurogi K, Murata H, Yoshida N, Ito M, Fukui M. Impact of metabolic phenotype changes on the development of chronic kidney disease: Panasonic cohort study 17. Obesity (Silver Spring) 2025. [PMID: 40265648 DOI: 10.1002/oby.24293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/23/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE This study investigated how changes in metabolic phenotype, defined by obesity and metabolic health, impact chronic kidney disease (CKD) development in a large cohort. METHODS A retrospective cohort study analyzed health data collected between 2011 and 2021 from 84,729 Panasonic Holdings Corporation (formerly Panasonic Corporation) employees aged ≥40 years. Metabolic phenotypes were classified as metabolically healthy with no obesity (MHNO), metabolically healthy with obesity (MHO), metabolic abnormalities with no obesity (MANO), and metabolic abnormalities with obesity (MAO). Changes in metabolic phenotype over 3 years and their association with CKD development were assessed using Cox proportional hazards models adjusted for confounding variables. RESULTS During a mean follow-up of 5.1 years, a total of 12,172 of participants (14.4%) developed CKD. Transitioning from MHO to MHNO and from MAO to MANO did not reduce CKD risk compared to each stable group. In contrast, participants in the MANO-to-MHNO and MAO-to-MHO groups significantly lowered CKD risk relative to each stable group, with hazard ratios of 0.86 (95% CI: 0.77-0.96) and 0.83 (95% CI: 0.67-1.02), respectively. The same results were observed when a rapid decline in renal function was used as the outcome. CONCLUSIONS The improvement of metabolic profile might outweigh weight reduction in CKD risk.
Collapse
Affiliation(s)
- Junya Hironaka
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Tetsuro Kusaba
- Department of Nephrology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Hanako Nakajima
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Kazushiro Kurogi
- Department of Health Care Center, Panasonic Health Insurance Organization, Moriguchi, Japan
| | - Hiroaki Murata
- Department of Orthopaedic Surgery, Matsushita Memorial Hospital, Moriguchi, Japan
| | - Naoki Yoshida
- Department of Health Care Center, Panasonic Health Insurance Organization, Moriguchi, Japan
| | - Masato Ito
- Department of Health Care Center, Panasonic Health Insurance Organization, Moriguchi, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
3
|
Niendorf T, Gladytz T, Cantow K, Millward JM, Waiczies S, Seeliger E. Magnetic resonance imaging of renal oxygenation. Nat Rev Nephrol 2025:10.1038/s41581-025-00956-z. [PMID: 40269325 DOI: 10.1038/s41581-025-00956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Renal hypoxia has a key role in the pathophysiology of many kidney diseases. MRI provides surrogate markers of oxygenation, offering a critical opportunity to detect renal hypoxia. However, studies that have assessed the diagnostic performance of oxygenation MRI for kidney disorders have provided inconsistent results because MRI metrics do not fully capture the complexity of renal oxygenation. Most oxygenation MRI studies are descriptive in nature and fail to detail the pathophysiological importance of the imaging findings. These limitations have restricted the clinical application of oxygenation MRI and the full potential of this technology to facilitate early diagnosis, risk prediction and treatment monitoring of kidney disease has not yet been realized. Understanding of the relationship between renal tissue oxygenation and MRI metrics, which is affected by kidney size, tubular volume fraction and renal blood volume fraction, and measurement of these factors using novel MR methods is imperative for correct physiological interpretation of renal MR oximetry findings. Next steps to enable the clinical adoption of MR oximetry should involve multidisciplinary collaboration to address standardization of acquisition and data analysis protocols and establish reference values of MRI metrics.
Collapse
Affiliation(s)
- Thoralf Niendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany.
- Experimental and Clinical Research Center, A joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| | - Thomas Gladytz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
- Experimental and Clinical Research Center, A joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kathleen Cantow
- Institute of Translational Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Jason M Millward
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
- Experimental and Clinical Research Center, A joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sonia Waiczies
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Ultrahigh Field Facility (B.U.F.F.), Berlin, Germany
- Experimental and Clinical Research Center, A joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Erdmann Seeliger
- Institute of Translational Physiology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| |
Collapse
|
4
|
Lin H, Pan J, Zhang J, He Y, Ge Y, Niu D, Han W, Han X, Li F, Bai X, Feng X, Lin L, Shen R, Su X, Qiao X. Intermedin protects peritubular capillaries by inhibiting eNOS uncoupling through AMPK/GTPCH-I/BH4 pathway and alleviate CKD following AKI. Free Radic Biol Med 2025; 234:72-85. [PMID: 40228707 DOI: 10.1016/j.freeradbiomed.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Even after recovery of kidney function following AKI, progression to CKD may still occur, characterized by a reduction in peritubular capillaries (PTC) and subsequent kidney fibrosis. Reactive oxygen species (ROS) from uncoupled eNOS are suspected to damage endothelial cells and cause PTC rarefaction observed in AKI-CKD. Intermedin (IMD) inhibits eNOS uncoupling by activating AMPK, but its impact on AKI-CKD transition remains unclear. METHODS We utilized IMD-deficient (IMD-/-) mice to explore its effects on AKI-CKD transition, PTC density, endothelial damage, and kidney ROS in a kidney ischemia/reperfusion injury (IRI) model. To elucidate its protective mechanism for PTCs, we subsequently investigated the effects of IMD on endothelial cells and ROS using a hypoxia/reoxygenation (HR) model with human umbilical vein endothelial cells (HUVECs). Finally, we investigated the influence of IMD on AMPK/GTPCH-I/BH4/eNOS to explore its mechanism in alleviating oxidative stress. RESULTS Compared with IMD+/+ littermate sham controls, PTC density was significantly reduced in IMD-/- sham mice, with significantly increased oxidative stress. Post-AKI, both IMD+/+ and IMD-/- mice demonstrated substantial declines in kidney function and histology, along with significant fibrosis, PTC reduction, and heightened oxidative stress. Moreover, the severity of kidney damage in IMD-/- mice following AKI was considerably more pronounced than in IMD+/+ mice. HR significantly induced eNOS uncoupling and oxidative stress in HUVECs. Treatment with IMD effectively inhibited eNOS uncoupling and ROS production, achieving levels comparable to the antioxidant N-acetylcysteine. The inhibitory effect of IMD on eNOS uncoupling was abrogated when L-NAME was introduced after HR. HR significantly impaired AMPK activation, which could be reversed by IMD. Additional experiments with inhibitors of GTPCH-I and AMPK, and exogenous BH4, confirmed that IMD protects endothelial cells by activating AMPK/GTPCH-I/BH4, thereby inhibiting eNOS uncoupling and ROS production. CONCLUSION We concluded that IMD inhibits AKI-CKD transition by protecting endothelial cells of PTC via AMPK/GTPCH-I/BH4/eNOS pathway.
Collapse
Affiliation(s)
- Hui Lin
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Juan Pan
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Junhua Zhang
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yuyin He
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yuan Ge
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Dan Niu
- Department of Pathology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Weixia Han
- Department of Pathology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaoli Han
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Fan Li
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaomei Bai
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xinyuan Feng
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ling Lin
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ruihua Shen
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaole Su
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xi Qiao
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China; Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China; Kidney Research Center of Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
5
|
Yang Y, Gong S, Zhou C, Xin W, Qin S, Yao M, Lan Q, Liao W, Zhao J, Huang Y. REST contributes to renal fibrosis through inducing mitochondrial energy metabolism imbalance in tubular epithelial cells. Cell Commun Signal 2025; 23:176. [PMID: 40200371 PMCID: PMC11980176 DOI: 10.1186/s12964-025-02166-3] [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: 11/20/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Renal fibrosis represents the final common pathological manifestation of chronic kidney disease (CKD), yet the underlying mechanism remains elusive, and there is still a lack of effective targeted therapeutic strategy. Although previous research indicated that repressor element 1-silencing transcription factor (REST) contributed to acute kidney injury (AKI) in renal tubular epithelial cells (RTECs), its specific contribution to renal fibrosis and associated mechanisms remains largely unexplored. METHODS Renal biopsies from CKD patients were collected to evaluate the expression of REST. Kidney-specific Rest conditional knockout (Cdh16-Cre/Restflox/flox) mice were generated and employed unilateral ureter obstruction (UUO) models to investigate the role of REST in renal fibrosis. RNA sequencing was performed to elucidate the mechanism. Mitochondrial function was evaluated by transmission electron microscopy (TEM), reactive oxygen species (ROS), oxygen consumption rates (OCR), extracellular acidifcation rate (ECAR) and adenosine triphosphate (ATP). The severity of renal fibrosis was assessed through Western blot, immunofluorescent staining and immumohistochemical staining. Bioinformatic prediction, dual luciferase reporter gene assay, point mutation and chromatin immunoprecipitation (ChIP) assay were utilized to clarify the molecular mechanism. RESULTS REST was significantly up-regulated in the kidney tissues from CKD patients, UUO-induced fibrotic mouse models and TGF-β1-incubated RTECs. Notably, kidney-specific knockout of Rest prominently alleviated renal fibrosis by improving mitochondrial energy metabolism and restoring fatty acid oxidation. Mechanically, REST disturbed mitochondrial energy metabolism through repressing the transcription of oxoglutarate dehydrogenase-like (OGDHL) via directly binding to its promotor region. Further, pharmacological inhibition of REST using the specific REST inhibitor, X5050, significantly ameliorated the progression of renal fibrosis both in vitro and in vivo. CONCLUSIONS Our explorations revealed the upregulation of REST in renal fibrosis disrupts mitochondrial energy metabolism through transcriptionally suppressing OGDHL, which may act as a promising therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Yingxian Yang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shuiqin Gong
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Chun Zhou
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wang Xin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shaozong Qin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Mengying Yao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Qigang Lan
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wenhao Liao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
6
|
Sasaki K, Masaki T. Epigenetic histone modifications in kidney disease and epigenetic memory. Clin Exp Nephrol 2025:10.1007/s10157-025-02668-x. [PMID: 40186651 DOI: 10.1007/s10157-025-02668-x] [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: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding RNAs, are influenced by environmental factors and play a central role in the progression and therapeutic targeting of kidney diseases, such as diabetic kidney disease (DKD), chronic kidney disease (CKD), and hypertension. These epigenetic changes are also preserved as cellular memory, with this "epigenetic memory" known to have long-term effects on such chronic diseases. Histone modifications are readily reversible epigenetic changes that regulate gene expression by altering chromatin structure or providing docking sites for transcriptional regulators. From a disease perspective, the involvement of epigenetics and "epigenetic memory" in DKD, CKD, senescence, and hypertension has been increasingly studied in recent years. Targeting epigenetic mechanisms is, thus, expected to offer novel therapeutic strategies for these diseases. Advances in treatment include histone deacetylase inhibitors and methyltransferase inhibitors, their applications of which have expanded from oncology to nephrology. However, challenges such as long-term toxicity and off-target effects remain significant. Further elucidation of kidney-specific epigenetic pathways and memory mechanisms may pave the way for precision epigenetic therapies, enabling the reversal of pathological epigenetic signatures and the mitigation of disease progression. CONCLUSION This review integrates recent advancements, highlighting functional evidence that histone modifications, particularly histone tail methylation, are involved in the pathogenesis of kidney diseases. It also emphasizes the translational significance of these findings, underlining the potential of epigenetics-based therapies to transform the management of kidney diseases.
Collapse
Affiliation(s)
- Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
7
|
Li L, Cai W, Zhang H, Tang J, Yang Y, Huang Y, Xi Q, Zhang R. Bergapten Ameliorates Renal Fibrosis by Inhibiting Ferroptosis. Phytother Res 2025; 39:1355-1371. [PMID: 39764683 DOI: 10.1002/ptr.8425] [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: 06/18/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 03/11/2025]
Abstract
Renal fibrosis is the most common pathway for the development of end-stage renal disease (ESRD) in various kidney diseases. Currently, the treatment options for renal fibrosis are limited. Ferroptosis is iron-mediated lipid peroxidation, triggered mainly by iron deposition and ROS generation. Notably, the kidney is the most sensitive of all tissues to iron-dependent ferroptosis, and the inhibition of ferroptosis is an effective therapeutic strategy for the treatment of kidney fibrosis. Nonetheless, the pathways involved in ferroptosis in renal fibrosis are still unclear. Bergapten, a natural coumarin derivative, is mainly found in bergapten essential oil, grapefruit juice, and other commonly used plants, and it has various pharmacological effects. However, the role that ferroptosis plays in renal fibrosis and the potential therapeutic benefits of bergapten remain unclear. In this study, we investigated the therapeutic effects of bergapten on renal fibrosis and its mechanisms. We investigated the anti-fibrotic effects of bergapten in in vivo and in vitro models of renal fibrosis. Initially, network pharmacological analysis was employed to predict the potential therapeutic impact of bergapten on renal fibrosis. We then explored the potential therapeutic role of bergapten in obstructive nephropathy, which is due to unilateral ureteral obstruction (UUO). Furthermore, RNA-Seq was conducted to investigate the possible mechanism of bergapten against renal fibrosis. Additionally, Bergapten demonstrated a significant improvement in TGF-β1-induced fibrosis and RSL3-induced renal tubular epithelial cell ferroptosis; these findings are consistent with those of the in vivo studies. Our findings indicate that bergapten is a potential treatment for renal fibrosis. Treatment with bergapten significantly reduced the expression of fibronectin and α-SMA in the damaged kidneys of UUO mice, thereby improving fibrosis. Meanwhile, bergapten protected against fibrosis caused by TGF-β1 and ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibitor RSL3. Significantly, bergapten therapy alleviated renal fibrosis by modulating ferroptosis. We found that bergapten inhibited PI3K phosphorylation and indirectly restored GPX4 expression. In conclusion, we have revealed the nephroprotective effect of bergapten, whose mechanism of action is related to the inhibition of ferroptosis, and it is expected that it will be developed as a therapeutic agent for the treatment of renal fibrosis. This study aims to explore the effect of bergapten on renal fibrosis ferroptosis. Collectively, these results demonstrate that bergapten is an inhibitor of ferroptosis and provides a new treatment strategy for diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Li Li
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenqian Cai
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hao Zhang
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiuren Tang
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongjie Yang
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Huang
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Xi
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
8
|
Wang S, Cheng Y, Zhang Z, Liu W, Ou M, Yin T, Meng Y, Ban H, Gu W, Meng X, Zhang L, Du Y. Association between obstructive sleep apnea and chronic kidney disease: A cross-sectional and Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41437. [PMID: 39928765 PMCID: PMC11812998 DOI: 10.1097/md.0000000000041437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/04/2025] [Accepted: 01/16/2025] [Indexed: 02/12/2025] Open
Abstract
Previous observational studies have shown that obstructive sleep apnea (OSA) was associated with chronic kidney disease(CKD). Early diagnosis of OSA usually helps better prevent the occurrence of CKD. This cross-sectional investigation was conducted using data from the National Health and Nutrition Examination Survey, which was carried out between 2007 to 2008 and 2015 to 2016. Logistic regression model was employed to assess the impact of OSA on CKD. We did a mediation analysis to assess how much of the effect of OSA on CKD was mediated through mediators. Additionally, Mendelian randomization (MR) analysis assessed the causal link between OSA and various measures of renal impairment and possible mediators: obesity, hypertension and type 2 diabetes mellitus. In the cross-sectional study, the results of unadjusted model showed that participants with OSA had a higher risk of CKD compared to non-OSA (OR = 1.14, 95% confidence intervals [CI]: 1.01-1.28, P < .05). In mediation analysis, the proportion of hypertension and obesity mediating the effect of OSA on CKD was 41.83% and 30.74%, respectively. Univariate MR analysis results showed that: genetically predicted OSA was associated with decreased estimated glomerular filtration ratecystatin c (eGFRcystatin c) level (OR = 0.997, 95% CI: 0.995-0.999, P < .05), increased blood urea nitrogen (BUN) levels (OR = 1.023, 95% CI: 1.008-1.038, P < .05), increased serum creatinine levels (OR = 1.010, 95% CI: 1.002-1.018, P < .05), increased serum cystatin C levels (OR = 1.015, 95% CI: 1.005-1.026, P < .05). Multivariable MR results showed that obesity mediated the causal effect of OSA on eGFRcystatin c, BUN levels and serum cystatin C levels. The cross-sectional study revealed a positive relationship between OSA and CKD, which was mediated by hypertension and obesity. The MR analysis suggest that OSA was associated with several measures of renal impairment, which was mediated by obesity. These findings may inform prevention and intervention strategies against CKD.
Collapse
Affiliation(s)
- Shaokang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yupei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhe Zhang
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mi Ou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Tianlong Yin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yalu Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haipeng Ban
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wenlong Gu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xianggang Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lili Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
9
|
Kasai T, Fujita K, Komatsu T, Ueno T, Kojima R, Hanaoka K, Urano Y. Development of a near-infrared fluorescent probe for the selective detection of severe hypoxia. RSC Chem Biol 2025:d4cb00243a. [PMID: 39911856 PMCID: PMC11791654 DOI: 10.1039/d4cb00243a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/24/2024] [Indexed: 02/07/2025] Open
Abstract
Severely hypoxic environments with oxygen concentrations around 1% are often found in serious diseases such as ischemia and cancer. However, existing near-infrared (NIR) fluorescent probes that can visualize hypoxia are also activated in mildly hypoxic environments (around 5% oxygen). Here, in order to selectively detect severe hypoxia, we used julolidine-based SiR (JSiR) as a NIR fluorophore and developed T-azoJSiR640 as a fluorescent probe. T-azoJSiR640 was able to detect severe hypoxia (around 1% oxygen concentration or less) in live cell imaging. Furthermore, the ischemic liver in a portal-vein-ligated mouse model was successfully visualized in vivo.
Collapse
Affiliation(s)
- Takafumi Kasai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Kyohhei Fujita
- Graduate School of Medicine, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, Keio University 1-5-30 Shibakoen, Minato-ku Tokyo 105-8512 Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
- Graduate School of Medicine, The University of Tokyo 7-3-1 Hongo, Bunkyo-ku Tokyo 113-0033 Japan
| |
Collapse
|
10
|
OKI C, UNO K, SASASE T, TSUTSUI T, SEKIGUCHI K, YAMAGUCHI A, MANDAI K, SHINOHARA M, SUGIMOTO M, MAEKAWA T, MIYAJIMA K, OHTA T. High-fat/high-sucrose diet-induced renal changes in obese diabetic mice: a comparison with db/db and KK-Ay mice. J Vet Med Sci 2025; 87:138-146. [PMID: 39662940 PMCID: PMC11830451 DOI: 10.1292/jvms.24-0313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Many genetic and environmental factors are involved in the development and progression of diabetic kidney disease (DKD), and its pathology shows various characteristics. Animal models of DKD play an important role in elucidating its pathogenesis and developing new therapies. In this study, we investigated the pathophysiological features of two DKD animal models: db/db mice (background of hyperglycemia) and KK-Ay mice (background of hyperinsulinemia). Male and female mice were fed a high-fat/high-sucrose (HFS) diet for eight weeks. Two mouse models fed the HFS diet showed increases in urinary protein, kidney weight, and glomerular size, but these changes were pronounced in KK-Ay mice. Pathological examination revealed tubulointerstitial fibrosis in KK-Ay mice fed the HFS diet, but not in db/db mice. In addition, fat accumulation was observed in the macula densa of db/db mice and in the glomeruli of KK-Ay mice fed with the HFS diet. In conclusion, an HFS diet exacerbates renal lesions with tubulointerstitial fibrosis in KK-Ay mice, and KK-Ay mice fed an HFS diet are expected to be useful as a DKD model.
Collapse
Affiliation(s)
- Chika OKI
- Biological/Pharmacological Research Laboratories, Takatsuki
Research Center, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka,
Japan
- Laboratory of Animal Physiology and Functional Anatomy,
Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kinuko UNO
- Laboratory of Animal Physiology and Functional Anatomy,
Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tomohiko SASASE
- Biological/Pharmacological Research Laboratories, Takatsuki
Research Center, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka,
Japan
- Laboratory of Animal Physiology and Functional Anatomy,
Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Takahiro TSUTSUI
- Laboratory of Animal Physiology and Functional Anatomy,
Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Keita SEKIGUCHI
- Department of Nutritional Science and Food Safety, Faculty
of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Ayane YAMAGUCHI
- Department of Nutritional Science and Food Safety, Faculty
of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Kouhei MANDAI
- Department of Nutritional Science and Food Safety, Faculty
of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | | | - Miki SUGIMOTO
- Laboratory of Animal Physiology and Functional Anatomy,
Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tatsuya MAEKAWA
- Department of Nutritional Science and Food Safety, Faculty
of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 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
|
11
|
Choi S, You J, Kim YJ, Lee HC, Park HP, Park CK, Oh H. High Intraoperative Serum Lactate Level is Associated with Acute Kidney Injury after Brain Tumor Resection. J Neurosurg Anesthesiol 2025; 37:55-63. [PMID: 38291797 DOI: 10.1097/ana.0000000000000954] [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: 07/25/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Postoperative acute kidney injury (AKI) is associated with poor clinical outcomes. Identification of risk factors for postoperative AKI is clinically important. Serum lactate can increase in situations of inadequate oxygen delivery and is widely used to assess a patient's clinical course. We investigated the association between intraoperative serum lactate levels and AKI after brain tumor resection. METHODS Demographics, medical and surgical history, tumor characteristics, surgery, anesthesia, preoperative and intraoperative blood test results, and postoperative clinical outcomes were retrospectively collected from 4131 patients who had undergone brain tumor resection. Patients were divided into high (n=1078) and low (n=3053) lactate groups based on an intraoperative maximum serum lactate level of 3.35 mmol/L. After propensity score matching, 1005 patients were included per group. AKI was diagnosed using the Kidney Disease Improving Global Outcomes criteria, based on serum creatinine levels within 7 days after surgery. RESULTS Postoperative AKI was observed in 53 (1.3%) patients and was more frequent in those with high lactate both before (3.2% [n=35] vs. 0.6% [n=18]; P < 0.001) and after (3.3% [n=33] vs. 0.6% [n=6]; P < 0.001) propensity score matching. Intraoperative predictors of postoperative AKI were maximum serum lactate levels > 3.35 mmol/L (odds ratio [95% confidence interval], 3.57 [1.45-8.74], P = 0.005), minimum blood pH (odds ratio per 1 unit, 0.01 [0.00-0.24], P = 0.004), minimum hematocrit (odds ratio per 1%, 0.91 [0.84-1.00], P = 0.037), and mean serum glucose levels > 200 mg/dL (odds ratio, 6.22 [1.75-22.16], P = 0.005). CONCLUSION High intraoperative serum lactate levels were associated with AKI after brain tumor resection.
Collapse
Affiliation(s)
| | - Jiwon You
- Department of Anesthesiology and Pain Medicine
| | | | | | | | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyongmin Oh
- Department of Anesthesiology and Pain Medicine
| |
Collapse
|
12
|
Balogh DB, Hodrea J, Saeed A, Cserhalmi M, Rozsahegyi A, Lakat T, Lenart L, Szabo AJ, Wagner LJ, Fekete A. Sigma-1 Receptor as a Novel Therapeutic Target in Diabetic Kidney Disease. Int J Mol Sci 2024; 25:13327. [PMID: 39769092 PMCID: PMC11679586 DOI: 10.3390/ijms252413327] [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: 10/31/2024] [Revised: 11/28/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease. Current treatments for DKD do not halt renal injury progression, highlighting an urgent need for therapies targeting key disease mechanisms. Our previous studies demonstrated that activating the Sigma-1 receptor (S1R) with fluvoxamine (FLU) protects against acute kidney injury by inhibiting inflammation and ameliorating the effect of hypoxia. Based on these, we hypothesized that FLU might exert a similar protective effect in DKD. Diabetes was induced in male Wistar rats using streptozotocin, followed by a seven-week FLU treatment. Metabolic and renal parameters were assessed along with a histological analysis of glomerular damage and fibrosis. The effects of FLU on inflammation, hypoxia, and fibrosis were tested in human proximal tubular cells and normal rat kidney fibroblasts. FLU improved renal function and reduced glomerular damage and tubulointerstitial fibrosis. It also mitigated inflammation by reducing TLR4, IL6, and NFKB1 expressions and moderated the cellular response to tubular hypoxia. Additionally, FLU suppressed TGF-β1-induced fibrotic processes and fibroblast transformation. These findings suggest that S1R activation can slow DKD progression and protect renal function by modulating critical inflammatory, hypoxic, and fibrotic pathways; therefore, it might serve as a promising novel drug target for preventing DKD.
Collapse
Affiliation(s)
- Dora B. Balogh
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Judit Hodrea
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Adar Saeed
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Marcell Cserhalmi
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Alexandra Rozsahegyi
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Tamas Lakat
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Lilla Lenart
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Attila J. Szabo
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| | - Laszlo J. Wagner
- Department of Surgery, Transplantation, and Gastroenterology, Semmelweis University, 1082 Budapest, Hungary;
| | - Andrea Fekete
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (J.H.); (A.S.); (M.C.); (A.R.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary;
| |
Collapse
|
13
|
Singh H, Singh R, Singh A, Singh H, Singh G, Kaur S, Singh B. Role of oxidative stress in diabetes-induced complications and their management with antioxidants. Arch Physiol Biochem 2024; 130:616-641. [PMID: 37571852 DOI: 10.1080/13813455.2023.2243651] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 08/13/2023]
Abstract
Diabetes mellitus (DM) is a huge global health issue and one of the most studied diseases, with a large global prevalence. Oxidative stress is a cytotoxic consequence of the excessive development of ROS and suppression of the antioxidant defense system for ROS elimination, which accelerates the progression of diabetes complications such as diabetic neuropathy, retinopathy, and nephropathy. Hyperglycaemia induced oxidative stress causes the activation of seven major pathways implicated in the pathogenesis of diabetic complications. These pathways increase the production of ROS and RNS, which contributes to dysregulated autophagy, gene expression changes, and the development of numerous pro-inflammatory mediators which may eventually lead to diabetic complications. This review will illustrate that oxidative stress plays a vital role in the pathogenesis of diabetic complications, and the use of antioxidants will help to reduce oxidative stress and thus may alleviate diabetic complications.
Collapse
Affiliation(s)
- Hasandeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Rajanpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Arshdeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Harshbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Gurpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Sarabjit Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| |
Collapse
|
14
|
Masuda T, Nagata D. Glomerular pressure and tubular oxygen supply: a critical dual target for renal protection. Hypertens Res 2024; 47:3330-3337. [PMID: 39397109 DOI: 10.1038/s41440-024-01944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
The primary treatment goal of chronic kidney disease (CKD) is preserving renal function and preventing its progression to end-stage renal disease. Glomerular hypertension and tubular hypoxia are critical risk factors in CKD progression. However, the renal hemodynamics make it difficult to avoid both factors due to the existence of peritubular capillaries that supply oxygen to the renal tubules downstream from the glomerulus through the efferent arteriole. In the treatment strategies for balancing glomerular pressure and tubular oxygen supply, afferent and efferent arterioles of the glomerulus determine glomerular filtration rate and blood flow to the peritubular capillaries. Therefore, sodium-glucose cotransporter 2 inhibitors and angiotensin receptor-neprilysin inhibitors as well as classical renin-angiotensin system inhibitors, which can change the diameter of afferent and/or efferent arterioles, are promising options for balancing this dual target and achieving renal protection. This review focuses on the clinical importance of glomerular pressure and tubular oxygen supply and proposes an effective treatment modality for this dual target.
Collapse
Affiliation(s)
- Takahiro Masuda
- Division of Nephrology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Daisuke Nagata
- Division of Nephrology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
15
|
Kohagura K, Zamami R, Oshiro N, Shinzato Y, Uesugi N. Heterogeneous afferent arteriolopathy: a key concept for understanding blood pressure-dependent renal damage. Hypertens Res 2024; 47:3383-3396. [PMID: 39379463 PMCID: PMC11618077 DOI: 10.1038/s41440-024-01916-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: 03/31/2024] [Revised: 07/02/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Abstract
Hypertension, aging, and other factors are associated with arteriosclerosis and arteriolosclerosis, primary morphological features of nephrosclerosis. Although such pathological changes are not invariably linked with renal decline but are prevalent across chronic kidney disease (CKD), understanding kidney damage progression is more pragmatic than precisely diagnosing nephrosclerosis itself. Hyalinosis and medial thickening of the afferent arteriole, along with intimal thickening of small arteries, can disrupt the autoregulatory system, jeopardizing glomerular perfusion pressure given systemic blood pressure (BP) fluctuations. Consequently, such vascular lesions cause glomerular damage by inducing glomerular hypertension and ischemia at the single nephron level. Thus, the interaction between systemic BP and afferent arteriolopathy markedly influences BP-dependent renal damage progression in nephrosclerosis. Both dilated and narrowed types of afferent arteriolopathy coexist throughout the kidney, with varying proportions among patients. Therefore, optimizing antihypertensive therapy to target either glomerular hypertension or ischemia is imperative. In recent years, clinical trials have indicated that combining renin-angiotensin system inhibitors (RASis) and sodium-glucose transporter 2 inhibitors (SGLT2is) is superior to using RASis alone in slowing renal function decline, despite comparable reductions in albuminuria. The superior efficacy of SGLT2is may arise from their beneficial effects on both glomerular hypertension and renal ischemia. A comprehensive understanding of the interaction between systemic BP and heterogeneous afferent arteriolopathy is pivotal for optimizing therapy and mitigating renal decline in patients with CKD of any etiology. Therefore, in this comprehensive review, we explore the role of afferent arteriolopathy in BP-dependent renal damage.
Collapse
Affiliation(s)
- Kentaro Kohagura
- Dialysis Unit, University of the Ryukyus Hospital, Okinawa, Japan.
| | - Ryo Zamami
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Nanako Oshiro
- Dialysis Unit, University of the Ryukyus Hospital, Okinawa, Japan
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yuki Shinzato
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Noriko Uesugi
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
16
|
Xu Y, Lu F, Wang M, Wang L, Ye C, Yang S, Wang C. Shen Shuai II recipe improves renal hypoxia to attenuate renal injury in 5/6 renal ablation/infarction rats and effect evaluation using blood oxygenation level-dependent functional magnetic resonance imaging. Ren Fail 2024; 46:2338565. [PMID: 38622926 PMCID: PMC11022919 DOI: 10.1080/0886022x.2024.2338565] [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: 11/22/2023] [Accepted: 03/31/2024] [Indexed: 04/17/2024] Open
Abstract
Background: Renal hypoxia plays a key role in the progression of chronic kidney disease (CKD). Shen Shuai II Recipe (SSR) has shown good results in the treatment of CKD as a common herbal formula. This study aimed to explore the effect of SSR on renal hypoxia and injury in CKD rats. Methods: Twenty-five Wistar rats underwent 5/6 renal ablation/infarction (A/I) surgery were randomly divided into three groups: 5/6 (A/I), 5/6 (A/I) + losartan (LOS), and 5/6 (A/I) + SSR groups. Another eight normal rats were used as the Sham group. After 8-week corresponding interventions, blood oxygenation level-dependent functional magnetic resonance imaging (BOLD-fMRI) was performed to evaluate renal oxygenation in all rats, and biochemical indicators were used to measure kidney and liver function, hemoglobin, and proteinuria. The expression of fibrosis and hypoxia-related proteins was analyzed using immunoblotting examination. Results: Renal oxygenation, evaluated by BOLD-fMRI as cortical and medullary T2* values (COT2* and MET2*), was decreased in 5/6 (A/I) rats, but increased after SSR treatment. SSR also downregulated the expression of hypoxia-inducible factor-1α (HIF-1α) in 5/6 (A/I) kidneys. With the improvement of renal hypoxia, renal function and fibrosis were improved in 5/6 (A/I) rats, accompanied by reduced proteinuria. Furthermore, the COT2* and MET2* were significantly positively correlated with the levels of creatinine clearance rate (Ccr) and hemoglobin, but negatively associated with the levels of serum creatinine (SCr), blood urea nitrogen (BUN), serum cystatin C (CysC), serum uric acid (UA), 24-h urinary protein (24-h Upr), and urinary albumin:creatinine ratio (UACR). Conclusion: The degree of renal oxygenation reduction is correlated with the severity of renal injury in CKD. SSR can improve renal hypoxia to attenuate renal injury in 5/6 (A/I) rats of CKD.
Collapse
Affiliation(s)
- Yizeng Xu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fang Lu
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingchen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chaoyang Ye
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Wang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Liang X, Jiang X. The hemoglobin, albumin, lymphocyte, and platelet (HALP) score is associated with severe renal tubular atrophy/interstitial fibrosis in IgA nephropathy. Eur J Med Res 2024; 29:542. [PMID: 39533443 PMCID: PMC11558843 DOI: 10.1186/s40001-024-02148-0] [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: 12/24/2023] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVES This study was performed to investigate the relationship between hemoglobin, albumin, lymphocyte and platelet (HALP) score and Oxford classification severe tubular atrophy/interstitial fibrosis (T2) in IgA nephropathy (IgAN). METHODS The clinical data and pathological findings of patients with IgA nephropathy diagnosed through renal biopsy at Hangzhou Hospital of Traditional Chinese Medicine between June 1, 2019 and May 31, 2022 were retrospectively collected and analyzed. The HALP score was calculated as hemoglobin (g/L) × albumin (g/L) × lymphocytes (/L)/ platelets (/L). According to the quartile of HALP scores in the study population, the subjects were divided into four groups: Quartile 1 (< 30.72), Quartile 2 (30.72-39.97), Quartile 3 (39.97-53.25) and Quartile 4(> 53.25). According to the extent of tubular atrophy/interstitial fibrosis, patients were categorized into mild to moderate injury (T0 + T1, ≤ 50%) and severe injury (T2, > 50%). The relationship between HALP score and severe tubular atrophy/interstitial fibrosis was investigated using Spearman's rank correlation coefficient, logistic regression analysis, restricted cubic splines (RCS), and receiver operating characteristic (ROC) curve analysis. RESULTS A total of 895 patients diagnosed with IgAN were included in this study, with an average age of 40.97 ± 12.261 years. Among them, 384 (42.9%) were male and 61 (6.8%) exhibited severe tubular atrophy/interstitial fibrosis. Multifactorial logistic regression analysis revealed that HALP independently influenced T2 (OR = 0.952, 95% CI 0.923-0.982, P = 0.002). Compared to Quartile 1, patients in Quartile 4 exhibited a significantly reduced risk of T2 (OR = 0.205, 95% CI 0.058-0.722, P = 0.014). Restricted cubic splines analysis revealed a linear inverse association between HALP and T2 risk (nonlinear P = 0.896). Furthermore, the receiver operating characteristic curve demonstrated that HALP possessed predictive value for T2 (AUC = 0.693, Jorden index = 0.324), and the cutoff value of HALP score is 36.54. CONCLUSIONS The risk of severe renal tubular atrophy/interstitial fibrosis is higher in IgAN patients with low HALP. HALP greater than 36.54 May reduce the risk of severe tubular atrophy/interstitial fibrosis.
Collapse
Affiliation(s)
- Xiaoli Liang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Stadium Road, Hangzhou, Zhejiang, 310000, People's Republic of China
| | - Xue Jiang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 453 Stadium Road, Hangzhou, Zhejiang, 310000, People's Republic of China.
| |
Collapse
|
18
|
Wu Z, Wang XR, Gao Y, Chen XH, Li M, Jin XF, He TT, Zhu YG, Chen XM, Zhou XH, Gao WJ. Study on the Correlation Between Renal Blood Perfusion and Kidney Injury in Different Weekly-Aged Type 2 Diabetic Mice. Physiol Res 2024; 73:717-727. [PMID: 39530907 PMCID: PMC11629956 DOI: 10.33549/physiolres.935405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/18/2024] [Indexed: 11/21/2024] Open
Abstract
This study aims to explore the correlation between renal blood perfusion (RBP) and diabetic nephropathy (DN). METHODS A total of 72 mice included db/db and db/m mice at the ages of 6, 14, and 22 weeks, forming six groups. RBP was assessed using Laser Speckle Contrast Imaging (LSCI). Kidney function markers and the extent of pathological damage were evaluated. Pearson correlation analysis was employed to predict the relationship between RBP and various indicators of kidney damage. RESULTS Compared to db/m mice of all ages, 6-week-old db/db mice showed no significant difference in kidney function markers and had no apparent pathological damage. However, db/db mice at other ages showed deteriorating kidney functions and evident pathological damage, which worsened with age. The RBP in db/m mice of all ages and 6-week-old db/db mice showed no significant difference; however, RBP in db/db mice demonstrated a significant declining trend with age. The correlation between RBP and kidney damage indicators was as follows: 24 h urinary microalbumin (r=-0.728), urinary transferrin (r=-0.834), urinary beta2-microglobulin (r=-0.755), urinary monocyte chemoattractant protein-1 (r=-0.786), Masson's trichrome staining (r=-0.872), and Periodic Acid-Schiff staining (r=-0.908). CONCLUSION RBP is strongly correlated with the extent of diabetic kidney damage.
Collapse
Affiliation(s)
- Z Wu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang, China. and ; National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
McLarnon SR, Honeycutt SE, N’Guetta PEY, Xiong Y, Li X, Abe K, Kitai H, Souma T, O’Brien LL. Altered renal vascular patterning reduces ischemic kidney injury and limits vascular loss associated with aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620969. [PMID: 39553980 PMCID: PMC11565873 DOI: 10.1101/2024.10.29.620969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The kidney vasculature has a complex arrangement, which runs in both series and parallel to perfuse the renal tissue and appropriately filter plasma. Recent studies have demonstrated that the development of this vascular pattern is dependent on netrin-1 secreted by renal stromal progenitors. Mice lacking netrin-1 develop an arterial tree with stochastic branching, particularly of the large interlobar vessels. The current study investigated whether abnormalities in renal vascular pattern altered kidney function or response to injury. To examine this, we analyzed kidney function at baseline as well as in response to recovery from a model of bilateral ischemic injury and measured vascular dynamics in aged mice. We found no differences in kidney function or morphology at baseline between mice with an abnormal arterial pattern compared to control. Interestingly, male and female mutant mice with stochastic vascular patterning showed a reduction in tubular injury in response to ischemia. Similarly, mutant mice also had a preservation of perfused vasculature with aging compared to a reduction in the control group. These results suggest that guided and organized patterning of the renal vasculature may not be required for normal kidney function; thus, modulating renal vascular patterning may represent an effective therapeutic strategy. Understanding how patterning and maturation of the arterial tree affects physiology and response to injury or aging has important implications for enhancing kidney regeneration and tissue engineering strategies.
Collapse
Affiliation(s)
- Sarah R. McLarnon
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Samuel E. Honeycutt
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Pierre-Emmanuel Y. N’Guetta
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yubin Xiong
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xinwei Li
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Cell and Developmental Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Koki Abe
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hiroki Kitai
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tomokazu Souma
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lori L. O’Brien
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
20
|
Giuliani KTK, Adams BC, Healy HG, Kassianos AJ. Regulated cell death in chronic kidney disease: current evidence and future clinical perspectives. Front Cell Dev Biol 2024; 12:1497460. [PMID: 39544363 PMCID: PMC11560912 DOI: 10.3389/fcell.2024.1497460] [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: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Chronic kidney disease (CKD) is the progressive loss of kidney function/structure over a period of at least 3 months. It is characterised histologically by the triad of cell loss, inflammation and fibrosis. This literature review focuses on the forms of cell death that trigger downstream inflammation and fibrosis, collectively called regulated cell death (RCD) pathways. Discrete forms of RCD have emerged as central mediators of CKD pathology. In particular, pathways of regulated necrosis - including mitochondrial permeability transition pore (mPTP)-mediated necrosis, necroptosis, ferroptosis and pyroptosis - have been shown to mediate kidney pathology directly or through the release of danger signals that trigger a pro-inflammatory response, further amplifying tissue injury in a cellular process called necroinflammation. Despite accumulating evidence in pre-clinical models, no clinical studies have yet targeted these RCD modes in human CKD. The review summarizes recent advances in our understanding of RCD pathways in CKD, looks at inter-relations between the pathways (with the emphasis on propagation of death signals) and the evidence for therapeutic targeting of molecules in the RCD pathways to prevent or treat CKD.
Collapse
Affiliation(s)
- Kurt T. K. Giuliani
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Benjamin C. Adams
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Helen G. Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
21
|
Ghalwash AA, El-Gohary RM, El Amrousy D, Morad LM, Kassem SS, Hegab II, Okasha AH. The gut microbiota metabolite trimethylamine-N-oxide in children with β-thalassemia: potential implication for iron-induced renal tubular dysfunction. Pediatr Res 2024:10.1038/s41390-024-03639-w. [PMID: 39448817 DOI: 10.1038/s41390-024-03639-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/22/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Renal tubular dysfunction is common in transfusion-dependent β thalassemia (β-TM). Iron overload, chronic anemia, and hypoxia are precipitating factors for renal insult. However, gut microbiota engagement in the renal insult has not been explored. Our work aimed to assess the potential link between iron overload, gut leakage/dysbiosis, and kidney dysfunction in these children. METHODS We enrolled 40 children with β-TM and 40 healthy controls. Gut leakage/dysbiosis biomarkers (trimethylamine-N-oxide [TMAO] and fecal short-chain fatty acids [SCFAs]), oxidative stress and inflammatory biomarkers, TMAO-regulated proteins such as serum sirtuin 1 (S.SIRT1) and serum high mobility box group-1 (S.HMGB1), and tubular dysfunction biomarkers were assessed. Correlations and regression analysis were performed to assess the relation between different parameters. RESULTS Iron overload, redox imbalance, and generalized inflammation were evident in children with β-TM. Renal tubular dysfunction biomarkers and S.TMAO were significantly elevated in the patient group. Furthermore, fecal SCFAs were significantly lower with upregulation of the investigated genes in the patient group. The correlation studies affirmed the close relationship between circulating ferritin, TMAO, and renal dysfunction and strongly implicated SIRT1/HMGB1 axis in TMAO action. CONCLUSIONS Gut dysbiosis may have a role in the pathogenesis of renal injury in children with β-TM. IMPACT Renal tubular dysfunction is a prominent health issue in β thalassemia major (β-TM). Iron overload, chronic anemia, and hypoxia are known precipitating factors. However, gut microbiota engagement in renal insult in these patients has not yet been explored. We aimed to assess potential link between iron overload, gut leakage/dysbiosis, and kidney dysfunction in β-TM children and to highlight the SIRT1/HMGB1 axis, a signal motivated by the gut microbiota-dependent metabolite trimethylamine-N-oxide (TMAO), involvement in such insults. We found that gut leakage/dysbiosis may have a role in kidney dysfunction in β-TM children by exacerbating the iron-motivated oxidative stress, inflammation, ferroptosis, and modulating SIRT1/HMGB1 axis.
Collapse
Affiliation(s)
- Asmaa A Ghalwash
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rehab M El-Gohary
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Doaa El Amrousy
- Pediatric Department, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Lamia M Morad
- Pediatric Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shaima S Kassem
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Asmaa H Okasha
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
22
|
Artan AS, Mirioğlu Ş, Hocaoğlu RH, Turgutalp K, Güllülü Boz SE, Eren N, Dinçer MT, Uzun S, Şahin G, Kutlay S, Cevher ŞK, Dheir H, Yılmaz M, Baştürk T, Tatar E, Kurultak İ, Öztürk R, Arıkan H, Yadigar S, Tunca O, Türkmen K, Elçioğlu ÖC, Kaya B, Karakan Ş, Ayar Y, Gül CB, Yazıcı H, Öztürk S. Observational study of immunosuppressive treatment patterns and outcomes in primary membranous nephropathy: a multicenter retrospective analysis. BMC Nephrol 2024; 25:327. [PMID: 39354386 PMCID: PMC11445947 DOI: 10.1186/s12882-024-03784-8] [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: 04/08/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND We evaluated the efficacy of different immunosuppressive regimens in patients with primary membranous nephropathy in a large national cohort. METHODS In this registry study, 558 patients from 47 centers who were treated with at least one immunosuppressive agent and had adequate follow-up data were included. Primary outcome was defined as complete (CR) or partial remission (PR). Secondary composite outcome was at least a 50% reduction in estimated glomerular filtration (eGFR), initiation of kidney replacement therapies, development of stage 5 chronic kidney disease, or death. RESULTS Median age at diagnosis was 48 (IQR: 37-57) years, and 358 (64.2%) were male. Patients were followed for a median of 24 (IQR: 12-60) months. Calcineurin inhibitors (CNIs) with or without glucocorticoids were the most commonly used regimen (43.4%), followed by glucocorticoids and cyclophosphamide (GC-CYC) (39.6%), glucocorticoid monotherapy (25.8%), and rituximab (RTX) (9.1%). Overall remission rate was 66.1% (CR 26.7%, PR 39.4%), and 59 (10.6%) patients reached secondary composite outcome. Multivariate logistic regression showed that baseline eGFR (OR 1.011, 95% CI: 1.003-1.019, p = 0.007), serum albumin (OR 1.682, 95% CI: 1.269-2.231, p < 0.001), and use of RTX (OR 0.296, 95% CI: 0.157-0.557, p < 0.001) were associated with remission rates; whereas only lower baseline hemoglobin was significantly associated with secondary composite outcome (OR: 0.843, 95% CI: 0.715-0.993, p = 0.041). CYC use was significantly associated with higher remission (OR 1.534, 95% CI: 1.027-2.290, p = 0.036). CONCLUSIONS Higher baseline eGFR and serum albumin levels correlated with increased remission rates. Remission rates were lower in patients treated with RTX, while those on GC-CYC showed higher rates of remission. Due to the study's retrospective nature and multiple treatments used, caution is warranted in interpreting these findings.
Collapse
Affiliation(s)
- Ayşe Serra Artan
- Department of Internal Medicine, Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey.
| | - Şafak Mirioğlu
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Bezmialem Vakıf University, Istanbul, Turkey
| | - Rabia Hacer Hocaoğlu
- Department of Internal Medicine, Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Kenan Turgutalp
- Department of Internal Medicine, Division of Nephrology, Mersin University School of Medicine, Mersin, Turkey
| | - Saide Elif Güllülü Boz
- Department of Internal Medicine, Division of Nephrology, Uludağ University Medical Faculty, Bursa, Turkey
| | - Necmi Eren
- Department of Internal Medicine, Division of Nephrology, Kocaeli University Medical Faculty, Kocaeli, Turkey
| | - Mevlüt Tamer Dinçer
- Department of Internal Medicine, Division of Nephrology, Istanbul University-Cerrahpaşa Cerrahpaşa Medical Faculty, Istanbul, Turkey
| | - Sami Uzun
- Division of Nephrology, University of Health Sciences, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Gülizar Şahin
- Division of Nephrology, University of Health Sciences S. Abdulhamid Research and Training Hospital, Istanbul, Turkey
| | - Sim Kutlay
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Ankara University, Ankara, Turkey
| | | | - Hamad Dheir
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Sakarya University, Sakarya, Turkey
| | - Mürvet Yılmaz
- Division of Nephrology, Bakırköy Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Taner Baştürk
- Division of Nephrology, University of Health Sciences Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Erhan Tatar
- Division of Nephrology, University of Health Sciences Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - İlhan Kurultak
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Trakya University, Edirne, Turkey
| | - Ramazan Öztürk
- Division of Nephrology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Hakkı Arıkan
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Marmara University, Istanbul, Turkey
| | - Serap Yadigar
- Division of Nephrology, Kartal Doktor Lütfi Kırdar City Hospital, Istanbul, Turkey
| | - Onur Tunca
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Afyonkarahisar Health Sciences University, Afyon, Turkey
| | - Kültigin Türkmen
- Department of Internal Medicine, Division of Nephrology, Necmettin Erbakan University Medical Faculty, Konya, Turkey
| | - Ömer Celal Elçioğlu
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Bezmialem Vakıf University, Istanbul, Turkey
| | - Bülent Kaya
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Çukurova University, Adana, Turkey
| | - Şebnem Karakan
- Faculty of Medicine, Department of Internal Medicine, Division of Nephrology, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Yavuz Ayar
- Faculty of Medicine Bursa City Health Application Research Center, Division of Nephrology, University of Health Sciences, Bursa, Turkey
| | - Cuma Bülent Gül
- Division of Nephrology, University of Health Sciences Bursa Faculty of Medicine, Bursa, Turkey
| | - Halil Yazıcı
- Department of Internal Medicine, Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Savaş Öztürk
- Department of Internal Medicine, Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
23
|
Zhou C, Ye Z, Zhang Y, He P, Liu M, Zhang Y, Yang S, Gan X, Nie J, Qin X. Association between lung function and risk of microvascular diseases in patients with diabetes: A prospective cohort and Mendelian randomization study. Nutr Metab Cardiovasc Dis 2024; 34:2378-2385. [PMID: 38862354 DOI: 10.1016/j.numecd.2024.05.004] [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: 02/25/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND AND AIMS To investigate causal relationships of lung function with risks microvascular diseases among participants with diabetes, type 2 diabetes mellitus (T2DM) and type 1 diabetes mellitus (T1DM), respectively, in prospective and Mendelian randomization (MR) study. METHODS AND RESULTS 14,617 participants with diabetes and without microvascular diseases at baseline from the UK Biobank were included in the prospective analysis. Of these, 13,421 had T2DM and 1196 had T1DM. The linear MR analyses were conducted in the UK Biobank with 6838 cases of microvascular diseases and 10,755 controls. Lung function measurements included forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1). The study outcome was microvascular diseases, a composite outcome including chronic kidney diseases, retinopathy and peripheral neuropathy. During a median follow-up of 12.1 years, 2668 new-onset microvascular diseases were recorded. FVC (%predicted) was inversely associated with the risk of new-onset microvascular diseases in participants with diabetes (Per SD increment, adjusted HR = 0.86; 95%CI:0.83-0.89), T2DM (Per SD increment, adjusted HR = 0.86; 95%CI:0.82-0.90) and T1DM (Per SD increment, adjusted HR = 0.87; 95%CI: 0.79-0.97), respectively. Similar results were found for FEV1 (%predicted). In MR analyses, genetically predicted FVC (adjusted RR = 0.55, 95%CI:0.39-0.77) and FEV1 (adjusted RR = 0.48, 95%CI:0.28-0.83) were both inversely associated with microvascular diseases in participants with T1DM. No significant association was found in those with T2DM. Similar findings were found for each component of microvascular diseases. CONCLUSION There was a causal inverse association between lung function and risks of microvascular diseases in participants with T1DM, but not in those with T2DM.
Collapse
Affiliation(s)
- Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yanjun Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Mengyi Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Sisi Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Xiaoqin Gan
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China
| | - Jing Nie
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China.
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China.
| |
Collapse
|
24
|
Smith JP, Paxton R, Medrano S, Sheffield NC, Sequeira-Lopez MLS, Ariel Gomez R. Inhibition of Renin Expression Is Regulated by an Epigenetic Switch From an Active to a Poised State. Hypertension 2024; 81:1869-1882. [PMID: 38989586 PMCID: PMC11337216 DOI: 10.1161/hypertensionaha.124.22886] [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: 02/13/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Renin-expressing cells are myoendocrine cells crucial for the maintenance of homeostasis. Renin is regulated by cAMP, p300 (histone acetyltransferase p300)/CBP (CREB-binding protein), and Brd4 (bromodomain-containing protein 4) proteins and associated pathways. However, the specific regulatory changes that occur following inhibition of these pathways are not clear. METHODS We treated As4.1 cells (tumoral cells derived from mouse juxtaglomerular cells that constitutively express renin) with 3 inhibitors that target different factors required for renin transcription: H-89-dihydrochloride, PKA (protein kinase A) inhibitor; JQ1, Brd4 bromodomain inhibitor; and A-485, p300/CBP inhibitor. We performed assay for transposase-accessible chromatin with sequencing (ATAC-seq), single-cell RNA sequencing, cleavage under targets and tagmentation (CUT&Tag), and chromatin immunoprecipitation sequencing for H3K27ac (acetylation of lysine 27 of the histone H3 protein) and p300 binding on biological replicates of treated and control As4.1 cells. RESULTS In response to each inhibitor, Ren1 expression was significantly reduced and reversible upon washout. Chromatin accessibility at the Ren1 locus did not markedly change but was globally reduced at distal elements. Inhibition of PKA led to significant reductions in H3K27ac and p300 binding specifically within the Ren1 super-enhancer region. Further, we identified enriched TF (transcription factor) motifs shared across each inhibitory treatment. Finally, we identified a set of 9 genes with putative roles across each of the 3 renin regulatory pathways and observed that each displayed differentially accessible chromatin, gene expression, H3K27ac, and p300 binding at their respective loci. CONCLUSIONS Inhibition of renin expression in cells that constitutively synthesize and release renin is regulated by an epigenetic switch from an active to poised state associated with decreased cell-cell communication and an epithelial-mesenchymal transition. This work highlights and helps define the factors necessary for renin cells to alternate between myoendocrine and contractile phenotypes.
Collapse
Affiliation(s)
- Jason P. Smith
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| | - Robert Paxton
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Silvia Medrano
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| | - Nathan C. Sheffield
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | | | - R. Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
25
|
Yamamoto T, Isaka Y. Pathological mechanisms of kidney disease in ageing. Nat Rev Nephrol 2024; 20:603-615. [PMID: 39025993 DOI: 10.1038/s41581-024-00868-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
The kidney is a metabolically active organ that requires energy to drive processes such as tubular reabsorption and secretion, and shows a decline in function with advancing age. Various molecular mechanisms, including genomic instability, telomere attrition, inflammation, autophagy, mitochondrial function, and changes to the sirtuin and Klotho signalling pathways, are recognized regulators of individual lifespan and pivotal factors that govern kidney ageing. Thus, mechanisms that contribute to ageing not only dictate renal outcomes but also exert a substantial influence over life expectancy. Conversely, kidney dysfunction, in the context of chronic kidney disease (CKD), precipitates an expedited ageing trajectory in individuals, leading to premature ageing and a disconnect between biological and chronological age. As CKD advances, age-related manifestations such as frailty become increasingly conspicuous. Hence, the pursuit of healthy ageing necessitates not only the management of age-related complications but also a comprehensive understanding of the processes and markers that underlie systemic ageing. Here, we examine the hallmarks of ageing, focusing on the mechanisms by which they affect kidney health and contribute to premature organ ageing. We also review diagnostic methodologies and interventions for premature ageing, with special consideration given to the potential of emerging therapeutic avenues to target age-related kidney diseases.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
26
|
Qiu X, Lan X, Li L, Chen H, Zhang N, Zheng X, Xie X. The role of perirenal adipose tissue deposition in chronic kidney disease progression: Mechanisms and therapeutic implications. Life Sci 2024; 352:122866. [PMID: 38936605 DOI: 10.1016/j.lfs.2024.122866] [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: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Chronic kidney disease (CKD) represents a significant and escalating global health challenge, with morbidity and mortality rates rising steadily. Evidence increasingly implicates perirenal adipose tissue (PRAT) deposition as a contributing factor in the pathogenesis of CKD. This review explores how PRAT deposition may exert deleterious effects on renal structure and function. The anatomical proximity of PRAT to the kidneys not only potentially causes mechanical compression but also leads to the dysregulated secretion of adipokines and inflammatory mediators, such as adiponectin, leptin, visfatin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and exosomes. Additionally, PRAT deposition may contribute to renal lipotoxicity through elevated levels of free fatty acids (FFA), triglycerides (TAG), diacylglycerol (DAG), and ceramides (Cer). PRAT deposition is also linked to the hyperactivation of the renin-angiotensin-aldosterone system (RAAS), which further exacerbates CKD progression. Recognizing PRAT deposition as an independent risk factor for CKD underscores the potential of targeting PRAT as a novel strategy for the prevention and management of CKD. This review further discusses interventions that could include measuring PRAT thickness to establish a baseline, managing metabolic risk factors that promote its deposition, and inhibiting key PRAT-induced signaling pathways.
Collapse
Affiliation(s)
- Xiang Qiu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Xin Lan
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Langhui Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Ningjuan Zhang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaoli Zheng
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
27
|
Mori K, Inoue T, Machiba Y, Uedono H, Nakatani S, Ishikawa M, Taniuchi S, Katayama Y, Yamamoto A, Kobayashi N, Kozawa E, Shimono T, Miki Y, Okada H, Emoto M. Effects of canagliflozin on kidney oxygenation evaluated using blood oxygenation level-dependent MRI in patients with type 2 diabetes. Front Endocrinol (Lausanne) 2024; 15:1451671. [PMID: 39280006 PMCID: PMC11393780 DOI: 10.3389/fendo.2024.1451671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024] Open
Abstract
Background Recent clinical studies suggest protective effects of SGLT2 inhibitors on kidney disease outcome. Chronic hypoxia has a critical role in kidney disease development, thus we speculated that canagliflozin, an SGLT2 inhibitor, can improve kidney oxygenation. Methods A single-arm study was conducted to investigate the effects of canagliflozin on T2* value, which reflects oxygenation level, in patients with type 2 diabetes (T2D) using repeated blood oxygenation level-dependent MRI (BOLD MRI) examinations. Changes in cortical T2* from before (Day 0) to after single-dose treatment (Day 1) and after five consecutive treatments (Day 5) were evaluated using 12-layer concentric objects (TLCO) and region of interest (ROI) methods. Results In the full analysis set (n=14 patients), the TLCO method showed no change of T2* with canagliflozin treatment, whereas the ROI method found that cortical T2* was significantly increased on Day 1 but not on Day 5. Sensitivity analysis using TLCO in 13 well-measured patients showed that canagliflozin significantly increased T2* on Day 1 with no change on Day 5, whereas a significant improvement in cortical T2* following canagliflozin treatment was found on both Day 1 and 5 using ROI. Conclusions Short-term canagliflozin treatment may improve cortical oxygenation and lead to better kidney outcomes in patients with T2D.
Collapse
Affiliation(s)
- Katsuhito Mori
- Department of Nephrology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Tsutomu Inoue
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yuri Machiba
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hideki Uedono
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Masahiro Ishikawa
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
- School of Clinical Engineering, Faculty of Health and Medical Care, Saitama Medical University, Saitama, Japan
| | - Satsuki Taniuchi
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yutaka Katayama
- Department of Radiology, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Akira Yamamoto
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Naoki Kobayashi
- School of Clinical Engineering, Faculty of Health and Medical Care, Saitama Medical University, Saitama, Japan
| | - Eito Kozawa
- Department of Radiology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Taro Shimono
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hirokazu Okada
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Masanori Emoto
- Department of Nephrology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
28
|
Wanner C, Nangaku M, Kraus BJ, Zinman B, Mattheus M, Hantel S, Schumacher M, Ohneberg K, Schmoor C, Inzucchi SE. How do SGLT2 inhibitors protect the kidney? A mediation analysis of the EMPA-REG OUTCOME trial. Nephrol Dial Transplant 2024; 39:1504-1513. [PMID: 38323492 PMCID: PMC11361804 DOI: 10.1093/ndt/gfae032] [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: 08/23/2023] [Indexed: 02/08/2024] Open
Abstract
INTRODUCTION Mechanisms underlying kidney benefits with sodium-glucose cotransporter-2 (SGLT2) inhibition in heart failure and/or type 2 diabetes (T2D) with established cardiovascular disease are currently unclear. METHODS We evaluated post hoc the factors mediating the effect of empagliflozin on a composite kidney outcome (first sustained estimated glomerular filtration rate ≥40% reduction from baseline, initiation of renal replacement therapy or death due to kidney disease) in EMPA-REG OUTCOME (Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients). Variables, calculated as change from baseline or updated mean, were evaluated as time-dependent covariates and using a landmark approach (at Week 12) in Cox regression analyses. In multivariable analyses, variables with the greatest mediating effect were added using a step-up procedure. RESULTS In univariable time-dependent updated mean covariate analyses, the strongest mediator was hematocrit (99.5% mediation). Hemoglobin, uric acid and urine albumin-to-creatinine ratio mediated 79.4%, 33.2% and 31.0%, respectively. Multivariable analyses were not performed due to the very strong mediation effect of hematocrit. In univariable Week 12 landmark change from baseline analyses, the strongest mediators included hematocrit (40.7%), glycated hemoglobin (28.3%), systolic blood pressure (16.8%) and free fatty acids (16.5%), which yielded a combined mediation of 78.9% in multivariable analysis. CONCLUSIONS Changes in hematocrit and hemoglobin were the strongest mediators of empagliflozin's kidney benefits in EMPA-REG OUTCOME participants with T2D and cardiovascular disease.
Collapse
Affiliation(s)
- Christoph Wanner
- Department of Medicine, Würzburg University Clinic, Würzburg, Germany
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Bettina J Kraus
- Medical Affairs, Boehringer Ingelheim International GmbH, Ingelheim, Germany
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Centre, University of Würzburg, Würzburg, Germany
| | - Bernard Zinman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Michaela Mattheus
- Biostatistics, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Stefan Hantel
- Biostatistics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Martin Schumacher
- Institute for Medical Biometry and Statistics and Clinical Trials Unit, Faculty of Medicine, and Medical Center, University of Freiburg, Freiburg, Germany
| | - Kristin Ohneberg
- Institute for Medical Biometry and Statistics and Clinical Trials Unit, Faculty of Medicine, and Medical Center, University of Freiburg, Freiburg, Germany
| | - Claudia Schmoor
- Clinical Trials Unit, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Silvio E Inzucchi
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
29
|
Mirioglu S, Cebeci E, Yazici H, Derici U, Sahin G, Coban G, Eren N, Gungor O, Dede F, Dincer T, Turkmen K, Basturk T, Duranay M, Arikan H, Tunca O, Elcioglu OC, Tatar E, Aydin Z, Oygar D, Demir S, Tanrisev M, Kurultak I, Oruc A, Turkmen A, Akcay OF, Cetinkaya H, Ozturk S. Prognostic factors and validation of the histologic chronicity score for C3 glomerulopathy: a registry analysis. Clin Kidney J 2024; 17:sfae077. [PMID: 39421234 PMCID: PMC11483614 DOI: 10.1093/ckj/sfae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Indexed: 10/19/2024] Open
Abstract
Background Data on the prognostic factors for C3 glomerulopathy (C3G) are limited, and validation of the new C3G histologic index (C3G-HI) in different settings is still needed. We aimed to evaluate the chronicity score of C3G-HI and probable prognostic factors in our population. Methods In this registry study, 74 patients from 20 centers with adequate follow-up data were included. Total chronicity score (TCS) was calculated according to percentages of glomerulosclerosis, interstitial fibrosis, tubular atrophy, and presence of arterio- and arteriolosclerosis. Primary composite outcome was defined as doubling of serum creatinine from baseline, undergoing dialysis or transplantation, development of stage 5 chronic kidney disease, or death. Results Median age was 34 [interquartile range (IQR) 24-46] years, and 39 patients (52.7%) were male. Median follow-up duration was 36 (IQR 12-60) months, and median TCS was 3 (IQR 1-5). Overall, 19 patients (25.7%) experienced primary composite outcome. Multivariate Cox regression model showed that only hemoglobin [adjusted HR (aHR) 0.67, 95% confidence interval 0.46-0.97, P = .035] predicted primary composite outcome, and TCS fell short of the statistical significance (aHR 1.26, 0.97-1.64, P = .08). Receiver operating characteristic analysis demonstrated that TCS showed an area under the curve value of 0.68 (0.56-0.78, P = .028) in discriminating primary composite outcome at 3 years, and 3-year kidney survival was lower in patients with TCS ≥4 (72.4%) compared with TCS <4 (91.1%) in Kaplan-Meier analysis (P = .036). Conclusions Low hemoglobin levels predicted dismal outcomes in patients with C3G. TCS ≥4 was associated with a worse 3-year kidney survival, which validated the 3-year prognostic value of the TCS of C3G-HI in our population.
Collapse
Affiliation(s)
- Safak Mirioglu
- Division of Nephrology, Bezmialem Vakif University Faculty of Medicine, Istanbul, Turkey
| | - Egemen Cebeci
- Division of Nephrology, Istanbul Haseki Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Halil Yazici
- Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Ulver Derici
- Division of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Gulizar Sahin
- Division of Nephrology, Sultan 2. Abdulhamid Han Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ganime Coban
- Department of Pathology, Bezmialem Vakif University Faculty of Medicine, Istanbul, Turkey
| | - Necmi Eren
- Division of Nephrology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Ozkan Gungor
- Division of Nephrology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaras, Turkey
| | - Fatih Dede
- Division of Nephology, Ankara Bilkent City Hospital, University of Health Sciences, Ankara, Turkey
| | - Tamer Dincer
- Division of Nephrology, Istanbul University-Cerrahpasa Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Kultigin Turkmen
- Division of Nephrology, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Taner Basturk
- Division of Nephrology, Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Murat Duranay
- Division of Nephrology, Ankara Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Hakki Arikan
- Division of Nephrology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Onur Tunca
- Division of Nephrology, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| | - Omer Celal Elcioglu
- Division of Nephrology, Bezmialem Vakif University Faculty of Medicine, Istanbul, Turkey
| | - Erhan Tatar
- Division of Nephrology, Bozyaka Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Zeki Aydin
- Division of Nephrology, Darica Farabi Training and Research Hospital, University of Health Sciences, Kocaeli, Turkey
| | - Deren Oygar
- Division of Nephrology, Dr Burhan Nalbantoglu State Hospital, Lefkosa, Cyprus
| | - Serap Demir
- Division of Nephrology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Mehmet Tanrisev
- Division of Nephrology, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Ilhan Kurultak
- Division of Nephrology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Aysegul Oruc
- Division of Nephrology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
| | - Aydin Turkmen
- Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Omer Faruk Akcay
- Division of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Hakki Cetinkaya
- Division of Nephrology, Sultan 2. Abdulhamid Han Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Savas Ozturk
- Division of Nephrology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
30
|
Tanaka N, Furukawa Y, Maeda T, Ishihara H, Dan K, Teramura M, Ichihashi K, Takase T, Takahashi Y, Tsuzura D, Shinoda A, Fujii M, Okada H, Itabashi F, Teramoto T. Renal ultrasonography predicts worsening renal function in patients with heart failure under tolvaptan administration. ESC Heart Fail 2024; 11:1911-1918. [PMID: 38468548 PMCID: PMC11287302 DOI: 10.1002/ehf2.14740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
AIMS Renal dysfunction in patients with chronic heart failure predicts a poor prognosis. Tolvaptan has a diuretic effect in patients with chronic kidney disease and heart failure without adverse effects on renal function. We aimed to determine the effects of tolvaptan and predictors of worsening renal function in patients with heart failure. METHODS AND RESULTS This post hoc analysis was a sub-analysis of a single-centre prospectively randomized trial on the early and short-term tolvaptan administration. We enrolled 201 participants with decompensated heart failure between January 2014 and March 2019 (early group, n = 104; age: 79.0 ± 12.8 years; late group, n = 97; age: 80.3 ± 10.8 years). Renal ultrasonography was performed before and after the administration of tolvaptan. Urine output and oral water intake significantly increased during tolvaptan administration. The difference between water intake and urine volume increased during tolvaptan administration. Changes in body weight, blood pressure, heart rate, and estimated glomerular filtration rate (eGFR) in both groups were comparable. The changes in peak-systolic velocity (PSV), acceleration time (AT) of the renal arteries, and resistance index were comparable. The changes in PSV and end-diastolic velocity (EDV) of the interlobar arteries increased following tolvaptan administration (Δmax PSV: 0.0 ± 14.8 cm/s before tolvaptan vs. 5.6 ± 15.7 cm/s after tolvaptan, P = 0.002; Δmean PSV: 0.4 ± 12.3 vs. 4.9 ± 12.7 cm/s, P = 0.002; Δmax EDV: -0.2 ± 3.5 vs. 1.4 ± 4.0 cm/s, P = 0.001; Δmean EDV: -0.0 ± 3.1 vs. 1.1 ± 3.4 cm/s, P = 0.003). The renal artery AT was negatively correlated with the eGFR (Δmax AT: beta = -0.2354, P = 0.044; Δmean AT: beta = -0.2477, P = 0.035). CONCLUSIONS Tolvaptan increased the PSV and EDV of the interlobar artery, which may mean tolvaptan increased renal blood flow. The renal artery AT may be a surrogate for worsening renal function.
Collapse
Affiliation(s)
- Nobukiyo Tanaka
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Yoshio Furukawa
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Takuya Maeda
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Hiroki Ishihara
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Kazuhiro Dan
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Masanori Teramura
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Kei Ichihashi
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Tetsuro Takase
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Yuya Takahashi
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Daichi Tsuzura
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Akira Shinoda
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Masato Fujii
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Hisashi Okada
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Fumiharu Itabashi
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| | - Tomohiko Teramoto
- Department of Cardiovascular MedicineIchinomiya Nishi Hospital1 Kaimei‐hiraIchinomiya494‐0001Japan
| |
Collapse
|
31
|
Luo H, Li J, Huang H, Jiao L, Zheng S, Ying Y, Li Q. AI-based segmentation of renal enhanced CT images for quantitative evaluate of chronic kidney disease. Sci Rep 2024; 14:16890. [PMID: 39043766 PMCID: PMC11266695 DOI: 10.1038/s41598-024-67658-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
To quantitatively evaluate chronic kidney disease (CKD), a deep convolutional neural network-based segmentation model was applied to renal enhanced computed tomography (CT) images. A retrospective analysis was conducted on a cohort of 100 individuals diagnosed with CKD and 90 individuals with healthy kidneys, who underwent contrast-enhanced CT scans of the kidneys or abdomen. Demographic and clinical data were collected from all participants. The study consisted of two distinct stages: firstly, the development and validation of a three-dimensional (3D) nnU-Net model for segmenting the arterial phase of renal enhanced CT scans; secondly, the utilization of the 3D nnU-Net model for quantitative evaluation of CKD. The 3D nnU-Net model achieved a mean Dice Similarity Coefficient (DSC) of 93.53% for renal parenchyma and 81.48% for renal cortex. Statistically significant differences were observed among different stages of renal function for renal parenchyma volume (VRP), renal cortex volume (VRC), renal medulla volume (VRM), the CT values of renal parenchyma (HuRP), the CT values of renal cortex (HuRC), and the CT values of renal medulla (HuRM) (F = 93.476, 144.918, 9.637, 170.533, 216.616, and 94.283; p < 0.001). Pearson correlation analysis revealed significant positive associations between glomerular filtration rate (eGFR) and VRP, VRC, VRM, HuRP, HuRC, and HuRM (r = 0.749, 0.818, 0.321, 0.819, 0.820, and 0.747, respectively, all p < 0.001). Similarly, a negative correlation was observed between serum creatinine (Scr) levels and VRP, VRC, VRM, HuRP, HuRC, and HuRM (r = - 0.759, - 0.777, - 0.420, - 0.762, - 0.771, and - 0.726, respectively, all p < 0.001). For predicting CKD in males, VRP had an area under the curve (AUC) of 0.726, p < 0.001; VRC, AUC 0.765, p < 0.001; VRM, AUC 0.578, p = 0.018; HuRP, AUC 0.912, p < 0.001; HuRC, AUC 0.952, p < 0.001; and HuRM, AUC 0.772, p < 0.001 in males. In females, VRP had an AUC of 0.813, p < 0.001; VRC, AUC 0.851, p < 0.001; VRM, AUC 0.623, p = 0.060; HuRP, AUC 0.904, p < 0.001; HuRC, AUC 0.934, p < 0.001; and HuRM, AUC 0.840, p < 0.001. The optimal cutoff values for predicting CKD in HuRP are 99.9 Hu for males and 98.4 Hu for females, while in HuRC are 120.1 Hu for males and 111.8 Hu for females. The kidney was effectively segmented by our AI-based 3D nnU-Net model for enhanced renal CT images. In terms of mild kidney injury, the CT values exhibited higher sensitivity compared to kidney volume. The correlation analysis revealed a stronger association between VRC, HuRP, and HuRC with renal function, while the association between VRP and HuRM was weaker, and the association between VRM was the weakest. Particularly, HuRP and HuRC demonstrated significant potential in predicting renal function. For diagnosing CKD, it is recommended to set the threshold values as follows: HuRP < 99.9 Hu and HuRC < 120.1 Hu in males, and HuRP < 98.4 Hu and HuRC < 111.8 Hu in females.
Collapse
Affiliation(s)
- Hui Luo
- Department of Radiology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Jingzhen Li
- Department of Nephrology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Haiyang Huang
- Department of Radiology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Lianghong Jiao
- Department of Radiology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Siyuan Zheng
- Department of Radiology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Yibo Ying
- Department of Radiology, Ningbo Yinzhou Second Hospital, Ningbo, China
| | - Qiang Li
- Department of Radiology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315000, China.
| |
Collapse
|
32
|
Nagashima R, Ishikawa H, Kuno Y, Kohda C, Eshima K, Iyoda M. Group2 innate lymphoid cells ameliorate renal fibrosis and dysfunction associated with adenine-induced CKD. Cell Immunol 2024; 401-402:104828. [PMID: 38759328 DOI: 10.1016/j.cellimm.2024.104828] [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: 01/23/2024] [Revised: 03/27/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Renal fibrosis is a common pathway of chronic kidney disease (CKD) progression involving primary kidney injury and kidney diseases. Group 2 innate lymphoid cells (ILC2s) mediate type 2 immune responses irrespective of antigen presentation and play a reno-protective role in kidney injury and disease. In the present study, we observed a decrease in kidney-resident ILC2s in CKD and found that enrichment of ILC2s in the kidney ameliorates renal fibrosis. In CKD kidney, ILC2s preferentially produced IL-13 over IL-5 in response to IL-33 stimulation, regardless of ST2L expression. Moreover, GATA3 expression was decreased in ILC2s, and T-bet+ ILC1s and RORγt+ ILC3s were increased in CKD kidney. Adoptive transfer of kidney ILC2s into adenine-induced CKD model mouse improved renal function and fibrosis. Renal fibroblasts cultured with IL33-activated kidney ILC2s suppressed myofibroblast trans-differentiation through Acta2 and Fn-1 regulation. These results suggest that kidney ILC2s prevent CKD progression via improvement of renal fibrosis. Our findings also suggest that ILC2s may contribute to the development of new therapeutic agents and strategies for tissue fibroses.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Immunology, Department of Biosciences, Kitasato University School of Science, Japan.
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan
| | - Koji Eshima
- Division of Immunology, Department of Biosciences, Kitasato University School of Science, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Japan
| |
Collapse
|
33
|
Wang L, Song Y, Zhang P, Chen W, Xiao F, Zhou P, Yang X, Dai H. Hypoxia-inducible factor prolyl hydroxylase inhibitor alleviates heatstroke-induced acute kidney injury by activating BNIP3-mediated mitophagy. FASEB J 2024; 38:e23723. [PMID: 38865198 DOI: 10.1096/fj.202400047r] [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: 01/08/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 06/14/2024]
Abstract
Hypoxia-induced inflammation and apoptosis are important pathophysiological features of heat stroke-induced acute kidney injury (HS-AKI). Hypoxia-inducible factor (HIF) is a key protein that regulates cell adaptation to hypoxia. HIF-prolyl hydroxylase inhibitor (HIF-PHI) stabilizes HIF to increase cell adaptation to hypoxia. Herein, we reported that HIF-PHI pretreatment significantly improved renal function, enhanced thermotolerance, and increased the survival rate of mice in the context of HS. Moreover, HIF-PHI could alleviate HS-induced mitochondrial damage, inflammation, and apoptosis in renal tubular epithelial cells (RTECs) by enhancing mitophagy in vitro and in vivo. By contrast, mitophagy inhibitors Mdivi-1, 3-MA, and Baf-A1 reversed the renoprotective effects of HIF-PHI. Mechanistically, HIF-PHI protects RTECs from inflammation and apoptosis by enhancing Bcl-2 adenovirus E18 19-kDa-interacting protein 3 (BNIP3)-mediated mitophagy, while genetic ablation of BNIP3 attenuated HIF-PHI-induced mitophagy and abolished HIF-PHI-mediated renal protection. Thus, our results indicated that HIF-PHI protects renal function by upregulating BNIP3-mediated mitophagy to improve HS-induced inflammation and apoptosis of RTECs, suggesting HIF-PHI as a promising therapeutic agent to treat HS-AKI.
Collapse
Affiliation(s)
- Ling Wang
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yongwei Song
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| | - Pan Zhang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Wenting Chen
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| | - Fei Xiao
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ping Zhou
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xuesen Yang
- Department of Tropical Medicine, Army Medical University, Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Huanzi Dai
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Hu W, Dai Y, Liu F, Yang T, Wang Y, Shen Y, Zhou W, Wu D, Gu L, Zhang M, Zhou Y. Assessing renal interstitial fibrosis using compartmental, non-compartmental, and model-free diffusion MRI approaches. Insights Imaging 2024; 15:156. [PMID: 38900336 PMCID: PMC11189852 DOI: 10.1186/s13244-024-01736-2] [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: 12/19/2023] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE To assess renal interstitial fibrosis (IF) using diffusion MRI approaches, and explore whether corticomedullary difference (CMD) of diffusion parameters, combination among MRI parameters, or combination with estimated glomerular filtration rate (eGFR) benefit IF evaluation. METHODS Forty-two patients with chronic kidney disease were included, undergoing MRI examinations. MRI parameters from apparent diffusion coefficient (ADC), intra-voxel incoherent motion (IVIM), diffusion kurtosis imaging (DKI), and diffusion-relaxation correlated spectrum imaging (DR-CSI) were obtained both for renal cortex and medulla. CMD of these parameters was calculated. Pathological IF scores (1-3) were obtained by biopsy. Patients were divided into mild (IF = 1, n = 23) and moderate-severe fibrosis (IF = 2-3, n = 19) groups. Group comparisons for MRI parameters were performed. Diagnostic performances were assessed by the receiver operator's curve analysis for discriminating mild from moderate-severe IF patients. RESULTS Significant inter-group differences existed for cortical ADC, IVIM-D, IVIM-f, DKI-MD, DR-CSI VB, and DR-CSI VC. Significant inter-group differences existed in ΔADC, ΔMD, ΔVB, ΔVC, ΔQB, and ΔQC. Among the cortical MRI parameters, VB displayed the highest AUC = 0.849, while ADC, f, and MD also showed AUC > 0.8. After combining cortical value and CMD, the diagnostic performances of the MRI parameters were slightly improved except for IVIM-D. Combining VB with f brings the best performance (AUC = 0.903) among MRI bi-variant models. A combination of cortical VB, ΔADC, and eGFR brought obvious improvement in diagnostic performance (AUC 0.963 vs 0.879, specificity 0.826 vs 0.896, and sensitivity 1.000 vs 0.842) than eGFR alone. CONCLUSION Our study shows promising results for the assessment of renal IF using diffusion MRI approaches. CRITICAL RELEVANCE STATEMENT Our study explores the non-invasive assessment of renal IF, an independent and effective predictor of renal outcomes, by comparing and combining diffusion MRI approaches including compartmental, non-compartmental, and model-free approaches. KEY POINTS Significant difference exists for diffusion parameters between mild and moderate-severe IF. Generally, cortical parameters show better performance than corresponding CMD. Bi-variant model lifts the diagnostic performance for assessing IF.
Collapse
Affiliation(s)
- Wentao Hu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongming Dai
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianshu Yang
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Wang
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Shen
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenyan Zhou
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongmei Wu
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronics Science, East China Normal University, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minfang Zhang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
Sohi GK, Farooqui N, Mohan A, Rajagopalan KS, Xing L, Zhu XY, Jordan K, Krier JD, Saadiq IM, Tang H, Hickson LJ, Eirin A, Lerman LO, Herrmann SM. The impact of hypoxia preconditioning on mesenchymal stem cells performance in hypertensive kidney disease. Stem Cell Res Ther 2024; 15:162. [PMID: 38853239 PMCID: PMC11163800 DOI: 10.1186/s13287-024-03778-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/27/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Autologous mesenchymal stem cells (MSCs) have emerged as a therapeutic option for many diseases. Hypertensive kidney disease (HKD) might impair MSCs' reparative ability by altering the biomolecular properties, but the characteristics of this impairment are unclear. In our previous pre-clinical studies, we found hypoxic preconditioning (HPC) enhanced angiogenesis and suppressed senescence gene expression. Thus, we hypothesize that HPC would improve human MSCs by enhancing their functionality and angiogenesis, creating an anti-inflammatory and anti-senescence environment. METHODS MSC samples (n = 12 each) were collected from the abdominal fat of healthy kidney donors (HC), hypertensive patients (HTN), and patients with hypertensive kidney disease (HKD). MSCs were harvested and cultured in Normoxic (20% O2) or Hypoxic (1% O2) conditions. MSC functionality was measured by proliferation assays and cytokine released in conditioned media. Senescence was evaluated by senescence-associated beta-galactosidase (SA-beta-gal) activity. Additionally, transcriptome analysis using RNA-sequencing and quantitative PCR (qPCR) were performed. RESULTS At baseline, normoxic HTN-MSCs had higher proliferation capacity compared to HC. However, HPC augmented proliferation in HC. HPC did not affect the release of pro-angiogenic protein VEGF, but increased EGF in HC-MSC, and decreased HGF in HC and HKD MSCs. Under HPC, SA-β-gal activity tended to decrease, particularly in HC group. HPC upregulated mostly the pro-angiogenic and inflammatory genes in HC and HKD and a few senescence genes in HKD. CONCLUSIONS HPC has a more favorable functional effect on HC- than on HKD-MSC, reflected in increased proliferation and EGF release, and modest decrease in senescence, whereas it has little effect on HTN or HKD MSCs.
Collapse
Affiliation(s)
- Gurparneet Kaur Sohi
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Naba Farooqui
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Arjunmohan Mohan
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | | | - Li Xing
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu province, China
| | - Xiang Y Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Kyra Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, 200, First Street SW, Rochester, 55902, MN, USA.
| |
Collapse
|
36
|
Tanaka S. Targeting inflammation in perivascular cells and neuroimmune interactions for treating kidney disease. Clin Exp Nephrol 2024; 28:505-512. [PMID: 38630367 PMCID: PMC11116252 DOI: 10.1007/s10157-024-02494-7] [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: 12/30/2023] [Accepted: 03/20/2024] [Indexed: 05/24/2024]
Abstract
Inflammation plays a crucial role in the pathophysiology of various kidney diseases. Kidney perivascular cells (pericytes/fibroblasts) are responsible for producing proinflammatory molecules, promoting immune cell infiltration, and enhancing inflammation. Vascular adhesion protein-1, expressed in kidney perivascular cells, is an ectoenzyme that catalyzes the oxidative deamination of primary amines with the production of hydrogen peroxide in the extracellular space. Our study demonstrated that blocking this enzyme suppressed hydrogen peroxide production and neutrophil infiltration, thereby reducing renal ischemia-reperfusion injury. Sphingosine 1-phosphate (S1P) signaling was also observed to play an essential role in the regulation of perivascular inflammation. S1P, which is produced in kidney perivascular cells, is transported into the extracellular space via spinster homolog 2, and then binds to S1P receptor-1 expressed in perivascular cells. Upon injury, inflammatory signaling in perivascular cells is enhanced by this pathway, thereby promoting immune cell infiltration and subsequent fibrosis. Furthermore, inhibition of S1P transport by spinster homolog 2 reduces kidney fibrosis. Hypoxia-inducible factor-prolyl hydroxylase inhibitors can restore the capacity for erythropoietin production in kidney perivascular cells. Animal data suggested that these drugs could also alleviate kidney and lipid inflammation although the precise mechanism is still unknown. Neuroimmune interactions have been attracting significant attention due to their potential to benefit patients with inflammatory diseases. Vagus nerve stimulation is one of the most promising strategies for harnessing neuroimmune interactions and attenuating inflammation associated with various diseases, including kidney disease. Using cutting-edge tools, the vagal afferents-C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis responsible for kidney protection induced by vagus nerve stimulation was identified in our study. Further research is required to decipher other crucial systems that control kidney inflammation and to determine whether these novel strategies can be applied to patients with kidney disease.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
37
|
Nakabayashi M, Tanabe J, Ogura Y, Ichinose M, Shibagaki Y, Kamijo-Ikemori A, Ono Y. Correlation of diabetic renal hypoperfusion with microvascular responses of the skeletal muscle: a rat model study using diffuse correlation spectroscopy. BIOMEDICAL OPTICS EXPRESS 2024; 15:3900-3913. [PMID: 38867789 PMCID: PMC11166419 DOI: 10.1364/boe.522385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 06/14/2024]
Abstract
Using diffuse correlation spectroscopy, we assessed the renal blood flow and thigh muscle microvascular responses in a rat model of type 2 diabetes. The blood flow index at the renal surface decreased significantly with arterial clamping, cardiac extirpation, and the progression of diabetic endothelial dysfunction. Renal blood flow measured in diabetic and nondiabetic rats also showed a significant correlation with the reactive hyperemic response of the thigh muscle. These results suggest shared microcirculatory dysfunction in the kidney and skeletal muscle and support endothelial responses in the skeletal muscle as a potential noninvasive biomarker of renal hypoperfusion.
Collapse
Affiliation(s)
- Mikie Nakabayashi
- Electrical Engineering Program, Graduate School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 2148571, Japan
| | - Jun Tanabe
- Division of Nephrology and hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 2168511, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 2168511, Japan
| | - Masashi Ichinose
- Human Integrative Physiology Laboratory, School of Business Administration, Meiji University, 1-1 Surugadai, Kanda, Chiyoda-ku Tokyo 1018301, Japan
| | - Yugo Shibagaki
- Division of Nephrology and hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 2168511, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 2168511, Japan
- Department of Anatomy, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 2168511, Japan
| | - Yumie Ono
- Department of Electronics and Bioinformatics, School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 2148571, Japan
| |
Collapse
|
38
|
Nangaku M. SGLT2 inhibitor: 2-way superstar in nephrology? Kidney Int 2024; 105:1176-1177. [PMID: 38777404 DOI: 10.1016/j.kint.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
39
|
Maeda S, Sakai S, Takabatake Y, Yamamoto T, Minami S, Nakamura J, Namba-Hamano T, Takahashi A, Matsuda J, Yonishi H, Matsui S, Imai A, Edahiro R, Yamamoto-Imoto H, Matsui I, Takashima S, Imamura R, Nonomura N, Yanagita M, Okada Y, Ballabio A, Nakamura S, Yoshimori T, Isaka Y. MondoA and AKI and AKI-to-CKD Transition. J Am Soc Nephrol 2024; 35:00001751-990000000-00338. [PMID: 38819935 PMCID: PMC11387036 DOI: 10.1681/asn.0000000000000414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/24/2024] [Indexed: 06/02/2024] Open
Abstract
Key Points
The expression of MondoA was decreased in the renal tubules of patients with CKD.Genetic ablation of MondoA in proximal tubules inhibited autophagy and increased vulnerability to AKI through increased expression of Rubicon.MondoA ablation during the recovery phase after ischemia-reperfusion aggravated kidney injury through downregulation of the transcription factor EB-peroxisome proliferator-activated receptor-γ coactivator-1α axis.
Background
Elderly individuals and patients with CKD are at a higher risk of AKI. The transcription factor MondoA is downregulated in the kidneys of aged individuals or patients with AKI; however, its roles in AKI development and the AKI-to-CKD transition remain unknown.
Methods
We investigated the expression of MondoA in human kidney biopsy samples, ischemia-reperfusion–injured (IRI) mouse kidneys, and cultured proximal tubular epithelial cells under hypoxia/reoxygenation. The role of MondoA during the initial and recovery phases after IRI was evaluated using proximal tubule–specific MondoA knockout mice and MondoA-deficient proximal tubular epithelial cells. Furthermore, we explored the involvement of Rubicon and transcription factor EB (TFEB), both of which are downstream factors of MondoA.
Results
MONDOA expression was decreased in the renal tubules of patients with CKD. In mouse kidneys, MondoA expression was decreased under ischemia, whereas its expression was increased during reperfusion. Genetic ablation of MondoA in proximal tubular epithelial cells inhibited autophagy and increased vulnerability to AKI through increased expression of Rubicon. Ablation of Rubicon in MondoA-deficient IRI kidneys activated autophagy and protected mitochondrial function. MondoA ablation during the recovery phase after ischemia-reperfusion aggravated kidney injury through downregulation of the TFEB-peroxisome proliferator-activated receptor-γ coactivator-1α axis. Pharmacological upregulation of TFEB contributed to maintaining mitochondrial biogenesis and increased peroxisome proliferator-activated receptor-γ coactivator-1α transcription.
Conclusions
Our findings demonstrate that MondoA protected against vulnerability to AKI by maintaining autophagy and subsequently supporting mitochondrial function to prevent progression to CKD.
Collapse
Grants
- JP22gm1410014 AMED
- 21K08276 a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology in Japan
- 22K16240 a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology in Japan
- 21H02935 a Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology in Japan
- None Novo Nordisk Pharma
- None Manpei Suzuki Diabetes Foundation
Collapse
Affiliation(s)
- Shihomi Maeda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinsuke Sakai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitsugu Takabatake
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Minami
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakamura
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Takahashi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Matsuda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Yonishi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sho Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsuhiro Imai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryoichi Imamura
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
| | - Shuhei Nakamura
- Department of Biochemistry, Nara Medical University, Nara, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
40
|
Zhang YY, Jin PP, Guo DZ, Bian D. Modified Zhenwu Tang delays chronic renal failure progression by modulating oxidative stress and hypoxic responses in renal proximal tubular epithelial cells. Heliyon 2024; 10:e31265. [PMID: 38803876 PMCID: PMC11128522 DOI: 10.1016/j.heliyon.2024.e31265] [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: 11/14/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Background Tubulointerstitial fibrosis (TIF) is a critical pathological feature of chronic renal failure (CRF), with oxidative stress (OS) and hypoxic responses in renal proximal tubular epithelial cells playing pivotal roles in disease progression. This study explores the effects of Modified Zhenwu Tang (MZWT) on these processes, aiming to uncover its potential mechanisms in slowing CRF progression. Methods We used adenine (Ade) to induce CRF in rats, which were then treated with benazepril hydrochloride (Lotensin) and MZWT for 8 weeks. Assessments included liver and renal function, electrolytes, blood lipids, renal tissue pathology, OS levels, the hypoxia-inducible factor (HIF) pathway, inflammatory markers, and other relevant indicators. In vitro, human renal cortical proximal tubular epithelial cells were subjected to hypoxia and lipopolysaccharide for 72 h, with concurrent treatment using MZWT, FM19G11, and N-acetyl-l-cysteine. Measurements taken included reactive oxygen species (ROS), HIF pathway activity, inflammatory markers, and other relevant indicators. Results Ade treatment induced significant disruptions in renal function, blood lipids, electrolytes, and tubulointerstitial architecture, alongside heightened OS, HIF pathway activation, and inflammatory responses in rats. In vivo, MZWT effectively ameliorated proteinuria, renal dysfunction, lipid and electrolyte imbalances, and renal tissue damage; it also suppressed OS, HIF pathway activation, epithelial-mesenchymal transition (EMT) in proximal tubular epithelial cells, and reduced the production of inflammatory cytokines and collagen fibers. In vitro findings demonstrated that MZWT decreased apoptosis, reduced ROS production, curbed OS, HIF pathway activation, and EMT in proximal tubular epithelial cells, and diminished the output of inflammatory cytokines and collagen. Conclusion OS and hypoxic responses significantly contribute to TIF development. MZWT mitigates these responses in renal proximal tubular epithelial cells, thereby delaying the progression of CRF.
Collapse
Affiliation(s)
- Yuan-yuan Zhang
- Graduate School, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, 050000, China
| | - Pei-pei Jin
- Hebei Yiling Hospital, Hebei, Shijiazhuang, 050000, China
| | - Deng-zhou Guo
- The First Affiliated Hospital of Hebei University of Chinese Medicine, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Chang'an District, Zhongshan East Road 389, 050011, China
| | - Dong Bian
- The First Affiliated Hospital of Hebei University of Chinese Medicine, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Chang'an District, Zhongshan East Road 389, 050011, China
| |
Collapse
|
41
|
Sun R, Li S, Wei Y, Hu L, Xu Q, Zhan G, Yan X, He Y, Wang Y, Li X, Luo A, Zhou Z. Development of interpretable machine learning models for prediction of acute kidney injury after noncardiac surgery: a retrospective cohort study. Int J Surg 2024; 110:2950-2962. [PMID: 38445452 PMCID: PMC11093510 DOI: 10.1097/js9.0000000000001237] [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: 12/01/2023] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Early identification of patients at high-risk of postoperative acute kidney injury (AKI) can facilitate the development of preventive approaches. This study aimed to develop prediction models for postoperative AKI in noncardiac surgery using machine learning algorithms. The authors also evaluated the predictive performance of models that included only preoperative variables or only important predictors. MATERIALS AND METHODS Adult patients undergoing noncardiac surgery were retrospectively included in the study (76 457 patients in the discovery cohort and 11 910 patients in the validation cohort). AKI was determined using the KDIGO criteria. The prediction model was developed using 87 variables (56 preoperative variables and 31 intraoperative variables). A variety of machine learning algorithms were employed to develop the model, including logistic regression, random forest, extreme gradient boosting, and gradient boosting decision trees. The performance of different models was compared using the area under the receiver operating characteristic curve (AUROC). Shapley Additive Explanations (SHAP) analysis was employed for model interpretation. RESULTS The patients in the discovery cohort had a median age of 52 years (IQR: 42-61 years), and 1179 patients (1.5%) developed AKI after surgery. The gradient boosting decision trees algorithm showed the best predictive performance using all available variables, or only preoperative variables. The AUROCs were 0.849 (95% CI: 0.835-0.863) and 0.828 (95% CI: 0.813-0.843), respectively. The SHAP analysis showed that age, surgical duration, preoperative serum creatinine, and gamma-glutamyltransferase, as well as American Society of Anesthesiologists physical status III were the most important five features. When gradually reducing the features, the AUROCs decreased from 0.852 (including the top 40 features) to 0.839 (including the top 10 features). In the validation cohort, the authors observed a similar pattern regarding the models' predictive performance. CONCLUSIONS The machine learning models the authors developed had satisfactory predictive performance for identifying high-risk postoperative AKI patients. Furthermore, the authors found that model performance was only slightly affected when only preoperative variables or only the most important predictive features were included.
Collapse
Affiliation(s)
- Rao Sun
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Yuna Wei
- Yidu Cloud Technology Inc, Beijing, People’s Republic of China
| | - Liu Hu
- Health Management Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Qiaoqiao Xu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Gaofeng Zhan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Xu Yan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Yuqin He
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Yao Wang
- Yidu Cloud Technology Inc, Beijing, People’s Republic of China
| | - Xinhua Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| | - Zhiqiang Zhou
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia
| |
Collapse
|
42
|
Yuan Y, Liang X, He M, Wu Y, Jiang X. Haemoglobin, albumin, lymphocyte, and platelet score as an independent predictor for renal prognosis in IgA nephropathy. Front Endocrinol (Lausanne) 2024; 15:1339921. [PMID: 38737556 PMCID: PMC11088234 DOI: 10.3389/fendo.2024.1339921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Objective The haemoglobin, albumin, lymphocyte, and platelet (HALP) score, a convenient and composite laboratory biomarker, can reflect inflammation and systemic nutritional status. This study was performed to investigate the effect of the HALP score on the prognosis of patients with IgA nephropathy (IgAN). Methods This is a retrospective single centre study that enrolled 895 biopsy-confirmed IgAN patients from June 2019 to June 2022 who were followed for more than 1 year. Kaplan-Meier curves and Cox regression analyses were performed to determine the relationship between HALP and adverse outcomes. The restricted cubic splines was used to identify the possible associations. The optimal cut-off value of HALP for renal poor outcome was identified by the area under the receiver operating characteristic curve (AUC). Results A total of 895 patients finally participated in the study and were divided into three groups (tertial 1-3) according to the baseline HALP score. More severe clinicopathologic features were observed in the lower HALP group, and Kaplan-Meier analysis showed patients in tertial 1 had a higher risk of kidney failure than the other groups (log-rank=11.02, P= 0.004). Multivariate Cox regression revealed that HALP score was an independent risk factor for renal prognosis in IgAN (adjusted HR: 0.967, 95% CI: 0.945-0.990, P = 0.006). The results of subgroup analysis suggested that HALP was more important in patients under the age of 50, BMI ≤ 23.9 and eGFR ≤ 90 mL/min/1.73 m2. The best cut-off HALP for renal survival was 38.83, sensitivity 72.1%, and specificity 55.9% (AUC: 0.662). Patients were further grouped according to HALP cut-off values and propensity matched. Multivariate Cox regression analysis revealed that HALP remained an independent predictor of IgAN in the matched cohort (HR 0.222, CI: 0.084-0.588, P=0.002). Conclusion HALP is a novel and potent composite parameter to predict kidney outcome in patients with IgAN.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Zhejiang Province, Management of Kidney Disease, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| | - Xiaoli Liang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Minhui He
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Wu
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue Jiang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Zhejiang Province, Management of Kidney Disease, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| |
Collapse
|
43
|
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
|
44
|
Silva BM, Macedo FH, Hayano EEM, Germano S, Ribeiro IF, Franco CA, Requião L, Medina-Pestana J, Goes MA. Relationship of hemoglobin levels with outcomes in deceased donor kidney transplant: a retrospective cohort study. J Bras Nefrol 2024; 46:e20230014. [PMID: 38284551 PMCID: PMC11210544 DOI: 10.1590/2175-8239-jbn-2023-0014en] [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: 02/03/2023] [Accepted: 11/03/2023] [Indexed: 01/30/2024] Open
Abstract
INTRODUCTION Anemia is frequent in patients undergoing replacement therapy for kidney failure. Anemia in the pre- and post-transplantation period might be related to kidney transplant outcomes. The current study therefore sought to assess the relationship between anemia, delayed allograft function (DGF), chronic kidney allograft dysfunction (CAD), and death from any cause following kidney transplantation from a deceased donor. METHODS This was a retrospective study with 206 kidney transplant patients of deceased donors. We analyzed deceased donors' and kidney transplant patients' demographic data. Moreover, we compared biochemical parameters, anemia status, and medicines between DGF and non-DGF groups. Afterward, we performed a multivariate analysis. We also evaluated outcomes, such as CAD within one year and death in ten years. RESULTS We observed a lower frequency of pre-transplant hemoglobin concentration (Hb) but higher frequency of donor-serum creatinine and red blood transfusion within one week after transplantation in the group with DGF. In addition, there was an independent association between Hb concentration before transplantation and DGF [OR 0.252, 95%CI: 0.159-0.401; p < 0.001]. There was also an association between Hb concentration after six months of kidney transplantation and both CAD [OR 0.798, 95% CI: 0.687-0.926; p = 0.003] and death from any cause. CONCLUSION An association was found between pre-transplantation anemia and DGF and between anemia six months after transplantation and both CAD and death by any cause. Thus, anemia before or after transplantation affects the outcomes for patients who have undergone kidney transplantation from a deceased donor.
Collapse
Affiliation(s)
| | | | | | - Suzeli Germano
- Universidade Federal de São Paulo, Divisão de Nefrologia, São Paulo,
SP, Brazil
| | | | - Carolina Azze Franco
- Universidade Federal de São Paulo, Departamento de Medicina, São
Paulo, SP, Brazil
| | - Lucio Requião
- Universidade Federal de São Paulo, Divisão de Nefrologia, São Paulo,
SP, Brazil
- Universidade Federal de São Paulo, Hospital do Rim, São Paulo, SP,
Brazil
| | - José Medina-Pestana
- Universidade Federal de São Paulo, Divisão de Nefrologia, São Paulo,
SP, Brazil
- Universidade Federal de São Paulo, Hospital do Rim, São Paulo, SP,
Brazil
| | - Miguel Angelo Goes
- Universidade Federal de São Paulo, Divisão de Nefrologia, São Paulo,
SP, Brazil
- Universidade Federal de São Paulo, Hospital do Rim, São Paulo, SP,
Brazil
| |
Collapse
|
45
|
Luo L, Zhang W, You S, Cui X, Tu H, Yi Q, Wu J, Liu O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol Res 2024; 202:107144. [PMID: 38484858 DOI: 10.1016/j.phrs.2024.107144] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Fibrosis is a pathological process that affects multiple organs and is considered one of the major causes of morbidity and mortality in multiple diseases, resulting in an enormous disease burden. Current studies have focused on fibroblasts and myofibroblasts, which directly lead to imbalance in generation and degradation of extracellular matrix (ECM). In recent years, an increasing number of studies have focused on the role of epithelial cells in fibrosis. In some cases, epithelial cells are first exposed to external physicochemical stimuli that may directly drive collagen accumulation in the mesenchyme. In other cases, the source of stimulation is mainly immune cells and some cytokines, and epithelial cells are similarly altered in the process. In this review, we will focus on the multiple dynamic alterations involved in epithelial cells after injury and during fibrogenesis, discuss the association among them, and summarize some therapies targeting changed epithelial cells. Especially, epithelial mesenchymal transition (EMT) is the key central step, which is closely linked to other biological behaviors. Meanwhile, we think studies on disruption of epithelial barrier, epithelial cell death and altered basal stem cell populations and stemness in fibrosis are not appreciated. We believe that therapies targeted epithelial cells can prevent the progress of fibrosis, but not reverse it. The epithelial cell targeting therapies will provide a wonderful preventive and delaying action.
Collapse
Affiliation(s)
- Liuyi Luo
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Oral Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siyao You
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Xinyan Cui
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Hua Tu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Qiao Yi
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China
| | - Jianjun Wu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| | - Ousheng Liu
- Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Matsuura T, Yoshimura A, Fukushima R. Effects of Beraprost with or without NOS Inhibition on Plasma Aldosterone and Hemodynamics in Healthy Cats. Vet Sci 2024; 11:155. [PMID: 38668422 PMCID: PMC11054574 DOI: 10.3390/vetsci11040155] [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: 02/02/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/29/2024] Open
Abstract
OBJECTIVES The aim of the study was to evaluate the hemodynamic and RA system effects of the oral administration of the clinical dose of beraprost for feline CKD in healthy cats, and also to examine whether NOS inhibition reversed them. METHODS A placebo-controlled pharmacological sequential design study was carried out to assess the plasma aldosterone and renin concentrations (PAC and PRC), blood pressure, heart rate, and exploratorily to estimate renal plasma flow (RPF) and renal vascular resistance (RVR) with simplified methods. RESULTS Beraprost reduced PAC when compared to the placebo (p < 0.05); this was reversed when NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME) was added to the beraprost treatment (p < 0.01). No differences in the PRC or hemodynamic parameters were detected between beraprost and the placebo. The correlation ratios (η2) showed opposite relationships between beraprost and the added L-NAME effects on PAC, mean blood pressure (MBP), heart rate, estimated RPF (p < 0.001), estimated RVR (p < 0.01), and PRC (p < 0.05). CONCLUSIONS In healthy cats, the clinical dose of beraprost suppresses PAC, which can be reversed by the inhibition of NOS.
Collapse
Affiliation(s)
| | - Aritada Yoshimura
- Animal Medical Center, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ryuji Fukushima
- Animal Medical Center, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
- Animal Medical Emergency Center, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan
| |
Collapse
|
47
|
Hao XM, Liu Y, Hailaiti D, Gong Y, Zhang XD, Yue BN, Liu JP, Wu XL, Yang KZ, Wang J, Liu QG. Mechanisms of inflammation modulation by different immune cells in hypertensive nephropathy. Front Immunol 2024; 15:1333170. [PMID: 38545112 PMCID: PMC10965702 DOI: 10.3389/fimmu.2024.1333170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Hypertensive nephropathy (HTN) is the second leading cause of end-stage renal disease (ESRD) and a chronic inflammatory disease. Persistent hypertension leads to lesions of intrarenal arterioles and arterioles, luminal stenosis, secondary ischemic renal parenchymal damage, and glomerulosclerosis, tubular atrophy, and interstitial fibrosis. Studying the pathogenesis of hypertensive nephropathy is a prerequisite for diagnosis and treatment. The main cause of HTN is poor long-term blood pressure control, but kidney damage is often accompanied by the occurrence of immune inflammation. Some studies have found that the activation of innate immunity, inflammation and acquired immunity is closely related to the pathogenesis of HTN, which can cause damage and dysfunction of target organs. There are more articles on the mechanism of diabetic nephropathy, while there are fewer studies related to immunity in hypertensive nephropathy. This article reviews the mechanisms by which several different immune cells and inflammatory cytokines regulate blood pressure and renal damage in HTN. It mainly focuses on immune cells, cytokines, and chemokines and inhibitors. However, further comprehensive and large-scale studies are needed to determine the role of these markers and provide effective protocols for clinical intervention and treatment.
Collapse
Affiliation(s)
- Xiao-Min Hao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | | | - Yu Gong
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Dong Zhang
- Department of Chinese Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Bing-Nan Yue
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ji-Peng Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Li Wu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ke-Zhen Yang
- Department of Rehabilitation Medicine, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing-Guo Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
48
|
Wang N, Zhang C. Recent Advances in the Management of Diabetic Kidney Disease: Slowing Progression. Int J Mol Sci 2024; 25:3086. [PMID: 38542060 PMCID: PMC10970506 DOI: 10.3390/ijms25063086] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 01/03/2025] Open
Abstract
Diabetic kidney disease (DKD) is a major cause of chronic kidney disease (CKD), and it heightens the risk of cardiovascular incidents. The pathogenesis of DKD is thought to involve hemodynamic, inflammatory, and metabolic factors that converge on the fibrotic pathway. Genetic predisposition and unhealthy lifestyle practices both play a significant role in the development and progression of DKD. In spite of the recent emergence of angiotensin receptors blockers (ARBs)/angiotensin converting enzyme inhibitor (ACEI), sodium-glucose cotransporter 2 (SGLT2) inhibitors, and nonsteroidal mineralocorticoid receptors antagonists (NS-MRAs), current therapies still fail to effectively arrest the progression of DKD. Glucagon-like peptide 1 receptor agonists (GLP-1RAs), a promising class of agents, possess the potential to act as renal protectors, effectively slowing the progression of DKD. Other agents, including pentoxifylline (PTF), selonsertib, and baricitinib hold great promise as potential therapies for DKD due to their anti-inflammatory and antifibrotic properties. Multidisciplinary treatment, encompassing lifestyle modifications and drug therapy, can effectively decelerate the progression of DKD. Based on the treatment of heart failure, it is recommended to use multiple drugs in combination rather than a single-use drug for the treatment of DKD. Unearthing the mechanisms underlying DKD is urgent to optimize the management of DKD. Inflammatory and fibrotic factors (including IL-1, MCP-1, MMP-9, CTGF, TNF-a and TGF-β1), along with lncRNAs, not only serve as diagnostic biomarkers, but also hold promise as therapeutic targets. In this review, we delve into the potential mechanisms and the current therapies of DKD. We also explore the additional value of combing these therapies to develop novel treatment strategies. Drawing from the current understanding of DKD pathogenesis, we propose HIF inhibitors, AGE inhibitors, and epigenetic modifications as promising therapeutic targets for the future.
Collapse
Affiliation(s)
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
49
|
Bjornstad P, Choi YJ, Platnick C, Gross S, Narongkiatikhun P, Melena I, Remmers L, Baca M, Schutte G, Dobbs T, Vigers T, Pyle L, Driscoll L, Tommerdahl K, Kendrick J, Looker HC, Dart A, Cherney D, van Raalte DH, Srivastava A, Li L, Prasad P, Saulnier P, Nelson RG, Johnson RJ, Nadeau KJ. Insulin Secretion, Sensitivity, and Kidney Function in Young Individuals With Type 2 Diabetes. Diabetes Care 2024; 47:409-417. [PMID: 38153805 PMCID: PMC10909687 DOI: 10.2337/dc23-1818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023]
Abstract
OBJECTIVE β-Cell dysfunction and insulin resistance magnify the risk of kidney injury in type 2 diabetes. The relationship between these factors and intraglomerular hemodynamics and kidney oxygen availability in youth with type 2 diabetes remains incompletely explored. RESEARCH DESIGN AND METHODS Fifty youth with type 2 diabetes (mean age ± SD 16 ± 2 years; diabetes duration 2.3 ± 1.8 years; 60% female; median HbA1c 6.4% [25th, 75th percentiles 5.9, 7.6%]; BMI 36.4 ± 7.4 kg/m2; urine albumin-to-creatinine ratio [UACR] 10.3 [5.9, 58.0] mg/g) 21 control participants with obesity (OCs; age 16 ± 2 years; 29% female; BMI 37.6 ± 7.4 kg/m2), and 20 control participants in the normal weight category (NWCs; age 17 ± 3 years; 70% female; BMI 22.5 ± 3.6 kg/m2) underwent iohexol and p-aminohippurate clearance to assess glomerular filtration rate (GFR) and renal plasma flow, kidney MRI for oxygenation, hyperglycemic clamp for insulin secretion (acute C-peptide response to glucose [ACPRg]) and disposition index (DI; ×103 mg/kg lean/min), and DXA for body composition. RESULTS Youth with type 2 diabetes exhibited lower DI (0.6 [0.0, 1.6] vs. 3.8 [2.4, 4.5] × 103 mg/kg lean/min; P < 0.0001) and ACPRg (0.6 [0.3, 1.4] vs. 5.3 [4.3, 6.9] nmol/L; P < 0.001) and higher UACR (10.3 [5.9, 58.0] vs. 5.3 [3.4, 14.3] mg/g; P = 0.003) and intraglomerular pressure (77.8 ± 11.5 vs. 64.8 ± 5.0 mmHg; P < 0.001) compared with OCs. Youth with type 2 diabetes and OCs had higher GFR and kidney oxygen availability (relative hyperoxia) than NWCs. DI was associated inversely with intraglomerular pressure and kidney hyperoxia. CONCLUSIONS Youth with type 2 diabetes demonstrated severe β-cell dysfunction that was associated with intraglomerular hypertension and kidney hyperoxia. Similar but attenuated findings were found in OCs.
Collapse
Affiliation(s)
- Petter Bjornstad
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Ye Ji Choi
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Carson Platnick
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Susan Gross
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Phoom Narongkiatikhun
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Isabella Melena
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Lauryn Remmers
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Madison Baca
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Grant Schutte
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Tyler Dobbs
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Tim Vigers
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Laura Pyle
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
| | - Lynette Driscoll
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Kalie Tommerdahl
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Jessica Kendrick
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Allison Dart
- Division of Nephrology, Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cherney
- Division of Nephrology, Department of Medicine, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Daniel H. van Raalte
- Diabetes Center, Department of Internal Medicine, Vrije Universiteit University Medical Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Anand Srivastava
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Luping Li
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL
| | - Pottumarthi Prasad
- Department of Radiology, NorthShore University HealthSystem, Evanston, IL
| | - Pierre Saulnier
- INSERM Centre d’Investigation Clinique 1402, CHU Poitiers, University of Poitiers, Poitiers, France
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Kristen J. Nadeau
- Section of Endocrinology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
50
|
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
|