1
|
Waheed YA, Liu J, Almayahe S, Sun D. The role of hyperuricemia in the progression of end-stage kidney disease and its molecular prospective in inflammation and cardiovascular diseases: A general review. Ther Apher Dial 2025. [PMID: 39966090 DOI: 10.1111/1744-9987.70000] [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: 11/04/2024] [Revised: 01/15/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
With the ongoing development of the Chinese economy, the occurrence of chronic kidney disease (CKD) has experienced a remarkable upsurge recently, and due to uremia caused by CKD, the number of patients undergoing dialysis has shown a dramatic increase. China has been ranked first in the world for patients undergoing hemodialysis (HD) and peritoneal dialysis (PD) with approximately one million patients across the country. Due to the loss of kidney function caused by CKD, the kidneys tend to lose their ability to excrete uric acid (UA) out of the body; therefore, most patients undergoing dialysis are complicated with hyperuricemia (HUA). HUA is an abnormal disease of purine metabolism, and it's considered a chronic disease. More than 90% of patients suffering from HUA will not show any symptoms on physical examination. According to statistics, if high serum UA is left untreated, 55% of patients will develop severe problems due to the purine crystallization in the body, and the kidneys are the most affected organs by HUA causing renal insufficiency that can promote end-stage kidney disease (ESKD) by activating the renin-angiotensin system (RAS), which will lead to inflammation, arteriosclerosis, cardiovascular diseases (CVD), and other diseases. Lifestyle modifications and pharmacological interventions are the first primary choice for lowering UA, although dialysis will tend to reduce the high UA levels in the blood, drugs are also necessary. This review will summarize the mechanisms and metabolism of UA, the relationship between HUA and ESKD progression, HUA and inflammation, HUA and CVD, and pharmacological treatment of HUA.
Collapse
Affiliation(s)
- Yousuf Abdulkarim Waheed
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Clinical Research Center for Kidney Disease Xuzhou Medical University, Xuzhou, China
| | - Jie Liu
- Department of Nephrology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Clinical Research Center for Kidney Disease Xuzhou Medical University, Xuzhou, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
2
|
Abdelrahman AM, Al Maskari R, Ali H, Manoj P, Al Suleimani Y. Effect of Riociguat on Adenine-Induced Chronic Kidney Disease in Rats. BIOLOGY 2025; 14:161. [PMID: 40001929 PMCID: PMC11851967 DOI: 10.3390/biology14020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025]
Abstract
Riociguat is a soluble guanylate cyclase (sGC) activator that increases the levels of cyclic guanosine monophosphate (cGMP). cGMP is known to play a key role in regulating kidney function. This research sought to investigate the possible protective effects of riociguat on the kidneys in the context of chronic kidney disease (CKD). CKD was induced in male Wistar rats through adenine administration. A total of 24 rats were allocated into four groups and administered treatments over a period of 35 days. Group 1 received a normal diet and a vehicle (carboxymethylcellulose (0.5%)), serving as the control. Group 2 received adenine (0.25% w/w) in the feed and a vehicle. Groups 3 and 4 received adenine in the feed (0.25% w/w) plus riociguat (3 mg/kg/day) and riociguat (10 mg/kg/day), respectively. Adenine administration significantly elevated systolic blood pressure, plasma creatinine, urea, and neutrophil gelatinase-associated lipocalin (NGAL). Furthermore, adenine reduced creatinine clearance and increased the urinary albumin-to-creatinine ratio and urinary N-Acetyl-β-D-Glucosaminidase (NAG). Histopathologically, adenine caused renal tubular necrosis and fibrosis. Furthermore, adenine elevated the plasma concentration of interleukins (IL-1β and IL-6) and tumor necrosis factor-alpha (TNF-α). Adenine significantly increased renal malondialdehyde (MDA) and reduced glutathione reductase (GR), superoxide dismutase (SOD), catalase (CAT), and total antioxidant capacity (TAC). Treatment with riociguat attenuated adenine-induced hypertension, improved kidney function, and ameliorated histopathological changes. Riociguat also reduced kidney injury markers, inflammation, and renal oxidative stress. The renoprotective effect of riociguat is probably due to anti-inflammatory and antioxidant actions. This indicates that riociguat may have the potential to slow the progression of kidney damage in chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Aly M. Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Raya Al Maskari
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman;
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| | - Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Muscat 123, Oman; (A.M.A.); (R.A.M.); (P.M.)
| |
Collapse
|
3
|
Ohata K, Sugaya T, Nguyen HN, Arai K, Hatanaka Y, Uno K, Tohma M, Uechi T, Sekiguchi K, Oikawa T, Nagabukuro H, Kuniyeda K, Kamijo-Ikemori A, Suzuki-Kemuriyama N, Nakae D, Noiri E, Miyajima K. Urinary liver-type fatty acid binding protein is a biomarker reflecting renal damage and the ameliorative effect of drugs at an early stage of histone-induced acute kidney injury. Nephrology (Carlton) 2024; 29:117-125. [PMID: 37950597 DOI: 10.1111/nep.14254] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
AIM Circulated histones play a crucial role in the pathogenesis of infectious diseases and severe trauma, and it is one of the potential molecular targets for therapeutics. Recently, we reported that histone is one of the causative agents for urinary L-FABP increase. However, the mechanism is still unclear, especially in severe cases. We further investigated the mechanism of urinary L-FABP increase using a more severe mouse model with histone-induced kidney injury. This study also aims to evaluate the therapeutic responsiveness of urinary L-FABP as a preliminary study. METHODS Human L-FABP chromosomal transgenic mice were administrated 30 mg/kg histone from a tail vein with a single dose. We also performed a comparative study in LPS administration model. For the evaluation of the therapeutic responsiveness of urinary L-FABP, we used heparin and rolipram. RESULTS The histological change with cast formation as a characteristic of the models was observed in proximal tubules. Urinary L-FABP levels were significantly elevated and these levels tended to be higher in those with more cast formation. Heparin and rolipram had the ameliorative effect of the cast formation induced by histone and urinary L-FABP levels significantly decreased. CONCLUSION Histone is one of the causative agents for the increase of urinary L-FABP at an early stage of AKI. In addition, it suggested that urinary L-FABP may be useful as a subclinical AKI marker reflecting kidney damage induced by histone. Furthermore, urinary L-FABP reflected the degree of the damage after the administration of therapeutic agents such as heparin and PDE4 inhibitor.
Collapse
Affiliation(s)
- Keiichi Ohata
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- CMIC Holdings Co., Ltd, Tokyo, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | - Hanh Nhung Nguyen
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Karin Arai
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuri Hatanaka
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Kinuko Uno
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Marika Tohma
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Teppei Uechi
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Keita Sekiguchi
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| | - Tsuyoshi Oikawa
- CMIC Holdings Co., Ltd, Tokyo, Japan
- Timewell Medical Co., Ltd, Tokyo, Japan
| | | | | | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Noriko Suzuki-Kemuriyama
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Dai Nakae
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
- Department of Medical Sports, Faculty of Health Care and Medical Sports, Teikyo Heisei University, Chiba, Japan
| | - Eisei Noiri
- National Center Biobank Network, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsuhiro Miyajima
- Department of Nutritional Science and Food Safety, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
- Department of Food and Nutritional Science, Graduate School of Agriculture, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
4
|
Zeng X, Liu Y, Fan Y, Wu D, Meng Y, Qin M. Agents for the Treatment of Gout: Current Advances and Future Perspectives. J Med Chem 2023; 66:14474-14493. [PMID: 37908076 DOI: 10.1021/acs.jmedchem.3c01710] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Gout is characterized by hyperuricemia and the deposition of monosodium urate (MSU) crystals around joints. Despite the availability of several drugs on the market, its treatment remains challenging owing to the notable side effects, such as hepatorenal toxicity and cardiovascular complications, that are associated with most existing agents. This perspective aims to summarize the current research progress in the development of antigout agents, particularly focusing on xanthine oxidase (XO) and urate anion transporter 1 (URAT1) inhibitors from a medicinal chemistry viewpoint and their preliminary structure-activity relationships (SARs). This perspective provides valuable insights and theoretical guidance to medicinal chemists for the discovery of antigout agents with novel chemical structures, better efficiency, and lower toxicity.
Collapse
Affiliation(s)
- Xiaoyi Zeng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuxin Fan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Di Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yangyang Meng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingze Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
5
|
Watanabe T, Ishikawa M, Abe K, Ishikawa T, Imakiire S, Masaki K, Hosokawa K, Fukuuchi T, Kaneko K, Ohtsubo T, Hirano M, Hirano K, Tsutsui H. Increased Lung Uric Acid Deteriorates Pulmonary Arterial Hypertension. J Am Heart Assoc 2021; 10:e022712. [PMID: 34845934 PMCID: PMC9075373 DOI: 10.1161/jaha.121.022712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Recent studies have demonstrated that uric acid (UA) enhances arginase activity, resulting in decreased NO in endothelial cells. However, the role of lung UA in pulmonary arterial hypertension (PAH) remains uncertain. We hypothesized that increased lung UA level contributes to the progression of PAH. Methods and Results In cultured human pulmonary arterial endothelial cells, voltage‐driven urate transporter 1 (URATv1) gene expression was detected, and treatment with UA increased arginase activity. In perfused lung preparations of VEGF receptor blocker (SU5416)/hypoxia/normoxia‐induced PAH model rats, addition of UA induced a greater pressure response than that seen in the control and decreased lung cGMP level. UA‐induced pressor responses were abolished by benzbromarone, a UA transporter inhibitor, or L‐norvaline, an arginase inhibitor. In PAH model rats, induction of hyperuricemia by administering 2% oxonic acid significantly increased lung UA level and induced greater elevation of right ventricular systolic pressure with exacerbation of occlusive neointimal lesions in small pulmonary arteries, compared with nonhyperuricemic PAH rats. Administration of benzbromarone to hyperuricemic PAH rats significantly reduced lung UA levels without changing XOR (xanthine oxidoreductase) activity, and attenuated right ventricular systolic pressure increase and occlusive lesion development. Topiroxostat, a XOR inhibitor, significantly reduced lung XOR activity in PAH rats, with no effects on increase in right ventricular systolic pressure, arterial elastance, and occlusive lesions. XOR‐knockout had no effects on right ventricular systolic pressure increase and arteriolar muscularization in hypoxia‐exposed mice. Conclusions Increased lung UA per se deteriorated PAH, whereas XOR had little impact. The mechanism of increased lung UA may be a novel therapeutic target for PAH complicated with hyperuricemia.
Collapse
Affiliation(s)
- Takanori Watanabe
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Mariko Ishikawa
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Anesthesiology and Critical Care MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Kohtaro Abe
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Tomohito Ishikawa
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Satomi Imakiire
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kohei Masaki
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kazuya Hosokawa
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
| | | | - Kiyoko Kaneko
- Faculty of Pharma‐ScienceTeikyo UniversityTokyoJapan
| | - Toshio Ohtsubo
- Department of Internal MedicineJapanese Red Cross Fukuoka HospitalFukuokaJapan
| | - Mayumi Hirano
- Division of Molecular CardiologyResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Cardiovascular PhysiologyFaculty of MedicineKagawa UniversityMiki‐cho, Kita‐gunKagawaJapan
| | - Katsuya Hirano
- Department of Cardiovascular PhysiologyFaculty of MedicineKagawa UniversityMiki‐cho, Kita‐gunKagawaJapan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular MedicineKyushu University Graduate School of Medical SciencesFukuokaJapan
- Division of Cardiovascular MedicineResearch Institute of AngiocardiologyGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
6
|
Angiotensin II type 1a receptor loss ameliorates chronic tubulointerstitial damage after renal ischemia reperfusion. Sci Rep 2021; 11:982. [PMID: 33441837 PMCID: PMC7806698 DOI: 10.1038/s41598-020-80209-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
We investigate whether suppressing the activation of the angiotensin II type 1a receptor (AT1a) can ameliorate severe chronic tubulointerstitial damage (TID) after renal ischemia reperfusion (IR) using AT1a knockout homozygous (AT1a−/−) male mice. To induce severe chronic TID after renal IR, unilateral renal ischemia was performed via clamping of the right renal pedicle in both AT1a−/− and wild-type (AT1a+/+) mice for 45 min. While marked renal atrophy and severe TID at 70 days postischemia was induced in the AT1a+/+ mice, such a development was not provoked in the AT1a−/− mice. Although the AT1a+/+ mice were administered hydralazine to maintain the same systolic blood pressure (SBP) levels as the AT1a−/− mice with lower SBP levels, hydralazine did not reproduce the renoprotective effects observed in the AT1a−/− mice. Acute tubular injury at 3 days postischemia was similar between the AT1a−/− mice and the AT1a+/+ mice. From our investigations using IR kidneys at 3, 14, and 28 days postischemia, the multiple molecular mechanisms may be related to prevention of severe chronic TID postischemia in the AT1a−/− mice. In conclusion, inactivation of the AT1 receptor may be useful in preventing the transition of acute kidney injury to chronic kidney disease.
Collapse
|
7
|
Feng X, Cao Y, Ding Y, Zheng H. Development and validation for the quantitative determination of xanthine oxidoreductase inhibitor topiroxostat by LC-MS/MS and its clinico-pharmacokinetic study. J Pharm Biomed Anal 2020; 189:113470. [DOI: 10.1016/j.jpba.2020.113470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022]
|
8
|
Jung SW, Kim SM, Kim YG, Lee SH, Moon JY. Uric acid and inflammation in kidney disease. Am J Physiol Renal Physiol 2020; 318:F1327-F1340. [PMID: 32223310 DOI: 10.1152/ajprenal.00272.2019] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Asymptomatic hyperuricemia is frequently observed in patients with kidney disease. Although a substantial number of epidemiologic studies have suggested that an elevated uric acid level plays a causative role in the development and progression of kidney disease, whether hyperuricemia is simply a result of decreased renal excretion of uric acid or is a contributor to kidney disease remains a matter of debate. Over the last two decades, multiple experimental studies have expanded the knowledge of the biological effects of uric acid beyond its role in gout. In particular, uric acid induces immune system activation and alters the characteristics of resident kidney cells, such as tubular epithelial cells, endothelial cells, and vascular smooth muscle cells, toward a proinflammatory and profibrotic state. These findings have led to an increased awareness of uric acid as a potential and modifiable risk factor in kidney disease. Here, we discuss the effects of uric acid on the immune system and subsequently review the effects of uric acid on the kidneys mainly in the context of inflammation.
Collapse
Affiliation(s)
- Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Su-Mi Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Tanabe J, Ogura Y, Kosaki K, Nagai Y, Sugaya T, Ohata K, Watanabe S, Ichikawa D, Inoue K, Hoshino S, Kimura K, Maeda S, Shibagaki Y, Kamijo-Ikemori A. Relationship between Urinary Liver-Type Fatty Acid-Binding Protein (L-FABP) and Sarcopenia in Spontaneously Diabetic Torii Fatty Rats. J Diabetes Res 2020; 2020:7614035. [PMID: 32405506 PMCID: PMC7201485 DOI: 10.1155/2020/7614035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/16/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a known risk factor for diabetic kidney disease (DKD) and sarcopenia in older patients. Because there may be an interaction between DKD and sarcopenia, the aim of the present study is to investigate the relationship between urinary levels of liver-type fatty acid-binding protein (L-FABP) and sarcopenia using a novel rat model of T2D. METHODS Male spontaneously diabetic Torii (SDT) fatty rats (n = 5) at 16 weeks of age were used as an animal model of T2D. Age- and sex-matched Sprague-Dawley (SD) rats (n = 7) were used as controls. Urine samples were obtained from the rats, and muscle strength was evaluated with the use of the forelimb grip test at 16, 20, and 24 weeks of age. Serum, kidney, soleus, and extensor digitorum longus (EDL) muscle samples were collected at 24 weeks of age. Urinary L-FABP levels were measured using dedicated enzyme-linked immunosorbent assays. RESULTS Increased urinary L-FABP levels, focal glomerular sclerosis, moderate interstitial inflammation and fibrosis, and accumulation of renal oxidative proteins were significantly observed in the SDT fatty rats, compared to the SD rats. Muscle weight, muscle strength, cross-sectional areas of both type I and type IIb muscle fibers, and increasing rate of muscle strength were significantly decreased in the SDT fatty rats compared to the SD rats at 24 weeks. Urinary L-FABP levels at 20 and 24 weeks were significantly negatively correlated with muscle strength. Urinary L-FABP levels at 16 weeks were significantly negatively correlated with the increasing rate of muscle strength. CONCLUSIONS Urinary L-FABP reflects the degree of muscle strength and weight, as well as cross-sectional areas of muscle fibers. Although further clinical study is needed, urinary L-FABP may be useful to monitor the progression of sarcopenia and DKD in T2D patients.
Collapse
Affiliation(s)
- Jun Tanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Keisei Kosaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yoshio Nagai
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Keiichi Ohata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Shiika Watanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuho Inoue
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Seiji Maeda
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| |
Collapse
|
10
|
Prediction of the pharmacokinetics and pharmacodynamics of topiroxostat in humans by integrating the physiologically based pharmacokinetic model with the drug-target residence time model. Biomed Pharmacother 2020; 121:109660. [DOI: 10.1016/j.biopha.2019.109660] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 01/12/2023] Open
|
11
|
Tanabe J, Ogura Y, Nakabayashi M, Nagai Y, Watanabe S, Sugaya T, Ohata K, Ichikawa D, Inoue K, Hoshino S, Kimura K, Shibagaki Y, Ono Y, Kamijo-Ikemori A. The Possibility of Urinary Liver-Type Fatty Acid-Binding Protein as a Biomarker of Renal Hypoxia in Spontaneously Diabetic Torii Fatty Rats. Kidney Blood Press Res 2019; 44:1476-1492. [PMID: 31734667 DOI: 10.1159/000503926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/04/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Renal hypoxia is an aggravating factor for tubulointerstitial damage, which is strongly associated with renal prognosis in diabetic kidney disease (DKD). Therefore, urinary markers that can detect renal hypoxia are useful for monitoring DKD. OBJECTIVE To determine the correlation between urinary liver-type fatty acid-binding protein (L-FABP) and renal hypoxia using a novel animal model of type 2 diabetes. METHODS Male spontaneously diabetic Torii (SDT) fatty rats (n = 6) were used as an animal model of type 2 diabetes. Age- and sex-matched Sprague-Dawley (SD) rats (n = 8) were used as controls. Body weight, systolic blood pressure, and blood glucose levels were measured at 8, 12, 16, and 24 weeks of age. Urine samples and serum and kidney tissues were collected at 24 weeks of age. Microvascular blood flow index (BFI) was measured using diffuse correlation spectroscopy before sampling both the serum and kidneys for the evaluation of renal microcirculation at the corticomedullary junction. RESULTS Obesity, hyperglycemia, and hypertension were observed in the SDT fatty rats. Focal glomerular sclerosis, moderate interstitial inflammation, and fibrosis were significantly more frequent in SDT fatty rats than in SD rats. While the frequency of peritubular endothelial cells and phosphoendothelial nitric oxide synthase levels were similar in both types of rats, the degree of renal hypoxia-inducible factor-1α (HIF-1α) expression was significantly higher (and with no change in renal vascular endothelial growth factor expression levels) in the SDT fatty rats. Urinary L-FABP levels were significantly higher and renal microvascular BFI was significantly lower in the SDT fatty rats than in the SD rats. Urinary L-FABP levels exhibited a significant positive correlation with renal HIF-1α expression and a significant negative correlation with renal microvascular BFI. CONCLUSIONS Urinary L-FABP levels reflect the degree of renal hypoxia in DKD in a type 2 diabetic animal model. Urinary L-FABP may thus prove useful as a renal hypoxia marker for monitoring DKD in patients with type 2 diabetes in clinical practice.
Collapse
Affiliation(s)
- Jun Tanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Mikie Nakabayashi
- Department of Electronics and Bioinformatics, School of Science and Technology, Meiji University, Kanagawa, Japan
| | - Yoshio Nagai
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Shiika Watanabe
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Keiichi Ohata
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Daisuke Ichikawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Kazuho Inoue
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Seiko Hoshino
- Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | - Yumie Ono
- Department of Electronics and Bioinformatics, School of Science and Technology, Meiji University, Kanagawa, Japan
| | - Atsuko Kamijo-Ikemori
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan, .,Department of Anatomy, St. Marianna University School of Medicine, Kanagawa, Japan,
| |
Collapse
|
12
|
Kosaki K, Sugaya T, Ohata K, Tanabe J, Hoshino S, Inoue K, Kimura K, Maeda S, Shibagaki Y, Kamijo-Ikemori A. Renoprotective effects of voluntary running exercise training on aldosterone-induced renal injury in human L-FABP chromosomal transgenic mice. Hypertens Res 2019; 42:1518-1527. [DOI: 10.1038/s41440-019-0273-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/08/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022]
|