1
|
Li AP, Zhang XX, Zhang QY, Wang MJ, Ju Z, Zhang XY, Qin XM, Liu GZ. Metabolomic profiling of adenine-induced CKD: pathway interconnections and kidney injury. Toxicol Res (Camb) 2025; 14:tfaf035. [PMID: 40135063 PMCID: PMC11932014 DOI: 10.1093/toxres/tfaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic kidney disease (CKD) is acknowledged as one of the largest public health problems in the world, characterized by a complex and diverse pathogenesis. Adenine-induced CKD, a classical model with multiple injury mechanisms, has been extensively employed in CKD research. However, the complete elucidation of the mechanisms underlying adenine-induced CKD remains elusive. In this study, the impacts of adenine (200 mg/kg/day) intake on the urine metabolome of rats were initially investigated using non-targeted metabolomics, and then targeted metabolomics was used to quantitatively verify key metabolites on crucial metabolic pathways. Interestingly, the interconnectedness of two significant pathways was discovered and validated through molecular biology techniques. The results found that adenine can cause significant perturbations in purine metabolism and the biosynthetic pathways of phenylalanine, tyrosine, and tryptophan. Subsequent targeted metabolomic analysis revealed a significant reduction in amino acid and hypoxanthine and creatinine levels in the kidneys of CKD rats, accompanied by an increase in xanthine level. Further analysis found that purine pathway can increase ROS production and affect the level of aromatic amino acid transporter SLC7A5, thus influencing the biosynthesis pathway of phenylalanine, tyrosine and tryptophan, ultimately contributing to kidney injury. This discovery provides offers novel insights into the underlying pathological mechanism of adenine-induced CKD. The development of chronic kidney disease is induced by multiple pathways of aromatic amino acid metabolism and purine metabolism.
Collapse
Affiliation(s)
- Ai-Ping Li
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
- Shanxi Traditional Chinese Medical Hospital, No. 46, Bingzhou West Street, Taiyuan 030012, China
| | - Xing-Xing Zhang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Qing-Yu Zhang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Meng-Jiao Wang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Zheng Ju
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Xiao-Yu Zhang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No. 92, Wucheng Road, Taiyuan 030006, Shanxi, China
| | - Guang-Zhen Liu
- Shanxi Traditional Chinese Medical Hospital, No. 46, Bingzhou West Street, Taiyuan 030012, China
| |
Collapse
|
2
|
Gao H, Xie T, Li Y, Xu Z, Song Z, Yu H, Zhou H, Li W, Yun C, Guan B, Luan S, Yin L. Role of gasdermins in chronic kidney disease. Front Immunol 2025; 16:1557707. [PMID: 40236694 PMCID: PMC11996640 DOI: 10.3389/fimmu.2025.1557707] [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: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Gasdermins (GSDMs), functioning as membrane perforating proteins, can be activated by canonical inflammasomes, noncanonical inflammasomes, as well as non-inflammasomes, leading to cell pyroptosis and the subsequent release of inflammatory mediators. Increasing evidence has implicated that GSDMs are associated with chronic kidney disease (CKD), including diabetes nephropathy, lupus nephritis, obstructive nephropathy, and crystalline nephropathy. This review centers on the role of GSDMs-mediated pyroptosis in the pathogenesis of CKD, providing novel ideas for enhancing the prognosis and therapeutic strategies of CKD.
Collapse
Affiliation(s)
- Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Ting Xie
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yunyi Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zigan Xu
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Zhuoheng Song
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Huixia Yu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Hongming Zhou
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Weilong Li
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Chen Yun
- Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Baozhang Guan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shaodong Luan
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Bódi B, Vágó RR, Nagy L, Ráduly AP, Gulyás A, Kupecz K, Azar L, Márványkövi FM, Szűcs G, Siska A, Cserni G, Földesi I, Papp Z, Sárközy M. Differential Myocardial Responses in Male and Female Rats with Uremic Cardiomyopathy. Int J Mol Sci 2025; 26:2259. [PMID: 40076880 PMCID: PMC11900185 DOI: 10.3390/ijms26052259] [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: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Uremic cardiomyopathy, characterized by diastolic dysfunction, left ventricular hypertrophy (LVH), and fibrosis, is a common cardiovascular complication of chronic kidney disease (CKD). Men are at a higher risk for cardiovascular and renal diseases, compared to age-matched, pre-menopausal women. We aimed to investigate the influence of sex on the severity of uremic cardiomyopathy through the characterization of functional and molecular indices of myocardial remodeling in a rat model. CKD was induced by a 5/6 nephrectomy in 9-week-old male and female Wistar rats. Serum and urine tests, transthoracic echocardiography, left ventricular (LV) histology, and quantitative reverse transcription polymerase chain reaction (RT-qPCR) were performed at week 8 or 9. Moreover, LV alterations were also tested in permeabilized cardiomyocytes (CMs) by force measurements and Western immunoblotting. CKD resulted in the development of a more severe uremic cardiomyopathy in male rats-including LVH, LV diastolic dysfunction, and fibrosis-than in female rats, where only LVH was observed. A uremic cardiomyopathy was also associated with a decrease in maximal Ca2+-activated force (Fmax) in CMs of male rats. Additionally, increases in CM Ca2+-independent passive stiffness (Fpassive) and decreases in cardiac myosin-binding protein C (cMyBP-C) phosphorylation levels were significantly larger in male than female rats. In conclusion, a uremic cardiomyopathy involved cardiac remodeling in both sexes. Nevertheless, male rats exhibited more pronounced signs of macroscopic and microscopic alterations than their female counterparts, illustrating a sex-dependent component of uremic cardiomyopathy.
Collapse
MESH Headings
- Animals
- Female
- Male
- Cardiomyopathies/etiology
- Cardiomyopathies/metabolism
- Cardiomyopathies/physiopathology
- Cardiomyopathies/pathology
- Uremia/metabolism
- Uremia/pathology
- Uremia/complications
- Rats
- Rats, Wistar
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/physiopathology
- Fibrosis
- Disease Models, Animal
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/pathology
- Sex Factors
- Echocardiography
- Carrier Proteins/metabolism
- Carrier Proteins/genetics
Collapse
Affiliation(s)
- Beáta Bódi
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (R.R.V.); (L.N.); (A.P.R.); (Z.P.)
| | - Rebeka Rita Vágó
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (R.R.V.); (L.N.); (A.P.R.); (Z.P.)
| | - László Nagy
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (R.R.V.); (L.N.); (A.P.R.); (Z.P.)
- Department of Cardiology, Division of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Arnold Péter Ráduly
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (R.R.V.); (L.N.); (A.P.R.); (Z.P.)
- Department of Cardiology, Division of Cardiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - András Gulyás
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.G.); (K.K.); (L.A.)
| | - Klaudia Kupecz
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.G.); (K.K.); (L.A.)
| | - Lilian Azar
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.G.); (K.K.); (L.A.)
| | - Fanni Magdolna Márványkövi
- Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (F.M.M.); (G.S.)
| | - Gergő Szűcs
- Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (F.M.M.); (G.S.)
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.S.); (I.F.)
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary;
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.S.); (I.F.)
| | - Zoltán Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (B.B.); (R.R.V.); (L.N.); (A.P.R.); (Z.P.)
| | - Márta Sárközy
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (A.G.); (K.K.); (L.A.)
- Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (F.M.M.); (G.S.)
| |
Collapse
|
4
|
Dunlap C, Zhao N, Ertl LS, Schall TJ, Sullivan KMC. C5aR expression in kidney tubules, macrophages and fibrosis. J Histotechnol 2025; 48:27-45. [PMID: 39607065 DOI: 10.1080/01478885.2024.2430041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The anaphylatoxin C5a and its receptor C5aR (CD88) are complement pathway effectors implicated in renal diseases, including ANCA-associated vasculitis. We investigated the kidney expression of C5aR and a second C5a receptor C5L2 by using immunohistochemistry, in situ hybridization, and spatial gene expression on formalin-fixed, paraffin-embedded human and mouse kidney. C5aR was detected on interstitial macrophages and in multiple tubular regions, including distal and proximal; C5L2 had a similar expression pattern. The 5/6 nephrectomy model of chronic kidney injury exhibited increased C5aR expression by infiltrating cells within the fibrotic regions. C5aR expression was confirmed on human leukocytes and in vitro differentiated macrophages by flow cytometry, and treatment with C5a induced the expression of chemokines and remodeling factors by macrophages, including CCL-3/-4/-7, -20, MMP-1/-3/-8/-12, and F3, and promoted leukocyte survival. C5a activity was C5aR dependent, as demonstrated by reversal with the C5aR inhibitor avacopan. Collectively, these results suggest that myeloid C5aR may induce excessive inflammation in the kidney via immune cell recruitment, extracellular matrix destruction, and remodeling, resulting in fibrotic tissue deposition.
Collapse
Affiliation(s)
- Carolyn Dunlap
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Niky Zhao
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Linda S Ertl
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | - Thomas J Schall
- Department of Biology, ChemoCentryx Inc., San Carlos, CA, USA
| | | |
Collapse
|
5
|
Schmiedt CW, Lourenço BN, Markovic LE, Lancaster M, Gumber S, Wannemacher J, Florian P, Coleman AE. Description of unilateral kidney embolism and contralateral nephrectomy as a less invasive remnant kidney model in cats; a proof-of-concept study. Animal Model Exp Med 2025. [PMID: 39956792 DOI: 10.1002/ame2.70001] [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: 10/07/2024] [Accepted: 01/18/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Refined models of kidney disease are critical to better understand disease processes and study novel treatments while minimizing discomfort in research animals. The objective of this study was to report a technique for minimally invasive partial kidney embolism in cats and describe outcomes following transcatheter administration of embolic microspheres with subsequent contralateral nephrectomy. METHODS Eleven, apparently healthy, male, purpose-bred cats underwent unilateral kidney embolism with 0.25 or 0.5 mL of embolic microparticle (40-120 μm) suspension (0.2 mL microspheres/mL) delivered into the right renal artery under fluoroscopic guidance, followed 5 months later by contralateral nephrectomy. One month after nephrectomy, blood and urinary markers of kidney function were evaluated, and embolized kidneys were harvested for histopathology evaluation. RESULTS Renal artery embolization was possible in all cats. Two cats did not complete the study, one after experiencing congestive heart failure (n = 1) and the other following evidence of complete kidney embolism precluding nephrectomy (n = 1) post-embolization. At study end, compared to baseline, cats had significant increases in median (range) serum creatinine (159.1 μmol/L [141.4-530.4] versus 128.2 μmol/L [92.8-150.3]; p = 0.0004), urea nitrogen (15.71 mmol/L [9.29-47.85] versus 7.50 mmol/L [6.07-8.57]; p < 0.0001), and symmetric dimethylarginine (0.74 μmol/L [0.59-3.12] versus 0.67 μmol/L [0.54-0.72]; p = 0.0288) concentrations. No differences in markers of kidney function were documented between dose groups. CONCLUSIONS Minimally invasive kidney embolism is a promising technique for modeling kidney disease in cats. Understanding optimal dose, timing of nephrectomy, and longer-term consequences requires additional work.
Collapse
Affiliation(s)
- Chad W Schmiedt
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Bianca N Lourenço
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Lauren E Markovic
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Meghan Lancaster
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Sanjeev Gumber
- Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | | | - Peter Florian
- Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | - Amanda E Coleman
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
6
|
Hamidizad Z, Kadkhodaee M, Kianian F, Ranjbaran M, Heidari F, Seifi B. Neuroprotective Effects of Sodium Nitroprusside on CKD-Induced Cognitive Dysfunction in Rats: Role of CBS and Nrf2/HO-1 Pathway. Neuromolecular Med 2025; 27:8. [PMID: 39775152 DOI: 10.1007/s12017-024-08828-8] [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: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Chronic kidney disease (CKD) is a conceivable new risk factor for cognitive disorder and dementia. Uremic toxicity, oxidative stress, and peripheral-central inflammation have been considered important mediators of CKD-induced nervous disorders. Nitric oxide (NO) is a retrograde neurotransmitter in synapses, and has vital roles in intracellular signaling in neurons. This research aims to determine the effectiveness of NO in CKD-induced cognitive deficits by considering the nuclear factor-erythroid factor 2-related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathway and the important roles of cystathionine beta-synthase (CBS, H2S producing enzyme). Forty rats were divided into four experimental groups: sham, five-sixth (5/6) nephrectomy (5/6Nx, CKD), CKD + NO donor (Sodium nitroprusside, SNP), CKD + SNP and a CBS inhibitor (amino-oxy acetic acid, AOAA). To assess the neurocognitive abilities, eleven weeks after 5/6Nx, behavioral tests (Novel object recognition test, Passive avoidance test, and Barnes maze test) were done. Twelfth week after 5/6Nx, blood urea nitrogen (BUN) and serum creatinine (sCr) levels, as well as the nuclear factor-erythroid factor 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) expression levels and neuronal injury in the hippocampus and prefrontal cortex were assessed. As predicted, the levels of BUN and sCr (both P < 0.001) and neuronal injury in the hippocampus (P < 0.001 for CA1; CA3; DG) and prefrontal cortex (P < 0.001) increased in CKD rats as well as 5/6Nx induced reduction of Nrf2 (both P < 0.001) /HO-1(P < 0.001; P < 0.01 respectively) pathway activity in the hippocampus and prefrontal cortex in CKD rats. Moreover, CKD leads to cognitive disorder and memory loss (Novel object recognition test (NOR) (P < 0.001), Passive avoidance test (PA) (P < 0.001) and Barnes maze (BA) (Escape latency (P < 0.001); Error (P < 0.001)). SNP treatment significantly improved Nrf2 (both P < 0.001) /HO-1 (P < 0.001; P < 0.05 respectively) pathways and neuronal injury (P < 0.001 for CA1; CA3; DG) in the hippocampus and prefrontal cortex in CKD rats as well as enhanced learning and memory ability in CKD rats. However, ameliorating effects of SNP on cognitive disorder (NOR (P < 0.05), PA (P < 0.001) and BA (Escape latency (P < 0.05); Error (P < 0.001)) and Nrf2 (P < 0.01; P < 0.001 in the hippocampus and prefrontal cortex respectively) /HO-1 (P < 0.05 in both) signaling pathway activity were nullified by CBS inhibitor and H2S reduction. In conclusion, this study demonstrated that NO improved CKD-induced cognitive impairment and neuronal death which is may be depended to CBS activity and endogenous H2S levels.
Collapse
Affiliation(s)
- Zeinab Hamidizad
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Farzaneh Kianian
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Fatemeh Heidari
- Department of Anatomy, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Behjat Seifi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran.
| |
Collapse
|
7
|
Li L, Zhao S, Liu X, Xu Z, Li D, Dai X. Lyophilized powder of calf bone marrow hydrolysate liposomes improved renal anemia: In vitro and in vivo evaluation. PLoS One 2024; 19:e0314811. [PMID: 39724079 DOI: 10.1371/journal.pone.0314811] [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: 07/30/2024] [Accepted: 11/16/2024] [Indexed: 12/28/2024] Open
Abstract
This study aimed to find whether oral administration of calf bone marrow hydrolysate liposomes (CBMHL) can improve renal anemia. Calf bone marrow was defatted, papain hydrolyzed, liposomalized and lyophilized. Its hematopoietic ability was proved by the colony formation experiment of umbilical cord blood hematopoietic stem cells in vitro. The rat model of renal anemia was established by adenine intragastric administration, and different concentrations of CBMHL were intragastricly administrated. Blood routine and serological indexes, transcription levels of hematopoietic factors and renal pathology were detected. From the appearance, redispersability, water content, liposome indexes and stability of Lyophilized powder of CBMHL, it could be concluded that the quality of freeze-dried CBMHL powder under this freeze-drying process was good. Compared with the control group, the burst forming unit-erythroid (BFU-E) in the CBMHL group was larger and the number of colonies increased significantly in the colony formation experiment (P < 0.05). The results of lyophilized powder of CBMHL co-culture with human adipose mesenchymal stem cells (MSCs) and human cytokine-induced killer (CIK) cells showed that the lyophilized powder of CBMHL had no potential toxicity and allergic reaction in vitro. Compared with the Model Group, the red blood cell (RBC) count, hemoglobin (HB) content and hematokrit (HCT) of rats blood routine in the Model+high doses of CBMHL Group (Model+H-CBMHL Group) increased significantly (P < 0.05). Serum erythropoietin (EPO) and glutathione (GSH) levels increased significantly (P < 0.05), while serum creatinine (Cr) levels decreased significantly(P < 0.05). The transcription level of Epo in kidney increased significantly (P < 0.05), the transcription levels of erythropoietin receptor (Epor) in bone marrow and interleukin 6 (Il6) in spleen were significantly increased (P < 0.01). The fragility of red blood cells decreased significantly, and the pathological structure of kidney improved significantly. It was proved that lyophilized powder of CBMHL could effectively enhance the hematopoietic ability of rats with renal anemia and protect the kidney structure and function.
Collapse
Affiliation(s)
- Li Li
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| | - Shasha Zhao
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| | - Xiaodun Liu
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| | - Zhe Xu
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| | - Dong Li
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| | - Xiaoyu Dai
- Department of Research and Development, Jinan Perfect Biological Technology Co., LTD, Jinan, Shandong, China
| |
Collapse
|
8
|
Vukovic A, Karanovic D, Mihailovic-Stanojevic ND, Miloradovic Z, Brkic P, Zivotic M, Nesovic Ostojic J, Ivanov M, Kovacevic S, Vajic UJ, Jovovic D, De Luka SR. Apocynin and Hyperbaric Oxygen Therapy Improve Renal Function and Structure in an Animal Model of CKD. Biomedicines 2024; 12:2788. [PMID: 39767695 PMCID: PMC11673868 DOI: 10.3390/biomedicines12122788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Chronic kidney disease (CKD) is a progressive pathological condition which results in the severe fibrosis of the kidneys. However, the mechanisms of CKD progression and fibrogenesis remain unclear. We wanted to examine the effects that apocynin and hyperbaric oxygen therapy (HBOT) have on renal function and structure in animals with CKD induced through 5/6 nephrectomy (5/6 Nx-L). METHODS Male Wistar rats were divided in 5 groups (n = 8/group) as follows: control-sham-operated rats; Nx-L-rats with 5/6 Nx-L; APO-5/6 Nx-L + apocynin treatment; HBOT-5/6 Nx-L + hyperbaric oxygen treatment, and APO+HBOT-5/6 Nx-L, treated with both treatments. All treatments started 4 weeks after the final step of CKD induction and lasted for 4 weeks. At the end of the experiment, urine samples were collected for the proteinuria assessment and the mean arterial pressure (MAP) was measured. Kidneys were collected for histopathological, Western blot, and immunohistochemical analyses. RESULTS All treatments significantly decreased MAP compared to the Nx-L group (p < 0.001). In the APO and APO+HBOT groups, the level of proteinuria was decreased compared to the Nx-L group (p < 0.05 and p < 0.01, respectively). All examined treatments significantly decreased the intensity of lesions in the kidney compared to those observed in the Nx-L group (p < 0.001). Isolated treatments with apocynin and HBOT induced a significant decrease in desmin expression compared to the Nx-L group (p < 0.05); meanwhile, they did not affect the levels of fibronectin (FN) and hypoxia-inducible factor-1α (HIF-1α). Combined treatment did not affect desmin expression levels; however, it induced a significant increase in fibronectin expression compared to Nx-L (p < 0.001). CONCLUSIONS Apocynin treatment decreased BP and protein loss, and it improved renal morphology at least partly through the downregulation of desmin expression without changing FN and HIF-1α. Hyperbaric oxygen therapy improved hypertension but failed to significantly affect the level of proteinuria. Combined treatment (apocynin and HBOT) normalized blood pressure (BP) values, renal function, and improved kidney structure by modulating FN and HIF-1α, without affecting desmin protein expression. Further studies are needed to elucidate the mechanisms of slowing down the progression of CKD in this experimental model.
Collapse
Affiliation(s)
- Andrija Vukovic
- Institute of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000 Belgrade, Serbia; (A.V.); (J.N.O.); (S.K.)
| | - Danijela Karanovic
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Nevena D Mihailovic-Stanojevic
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Zoran Miloradovic
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Predrag Brkic
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia;
| | - Maja Zivotic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000 Belgrade, Serbia;
| | - Jelena Nesovic Ostojic
- Institute of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000 Belgrade, Serbia; (A.V.); (J.N.O.); (S.K.)
| | - Milan Ivanov
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Sanjin Kovacevic
- Institute of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000 Belgrade, Serbia; (A.V.); (J.N.O.); (S.K.)
| | - Una-Jovana Vajic
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Djurdjica Jovovic
- Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, 11000 Belgrade, Serbia; (D.K.); (N.D.M.-S.); (Z.M.); (M.I.); (U.-J.V.); (D.J.)
| | - Silvio R. De Luka
- Institute of Pathological Physiology, Faculty of Medicine, University of Belgrade, Dr Subotića 1, 11000 Belgrade, Serbia; (A.V.); (J.N.O.); (S.K.)
| |
Collapse
|
9
|
Sun K, Qiu Y, Sun Y. Correlation analysis of AVPR1a and AVPR2 with abnormal water and sodium and potassium metabolism in rats. Open Life Sci 2024; 19:20220949. [PMID: 39588122 PMCID: PMC11588004 DOI: 10.1515/biol-2022-0949] [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: 03/04/2024] [Revised: 07/17/2024] [Accepted: 08/09/2024] [Indexed: 11/27/2024] Open
Abstract
In clinical practice, an increasing number of patients exhibit concurrent cardiac and renal dysfunction, known as "cardiorenal syndrome," where each condition exacerbates the other, resulting in poorer patient prognosis. Fluid and sodium retention can lead to excessive fluid overload in the body; therefore, correcting fluid and sodium metabolic disorders is crucial for alleviating patient symptoms. This study was to investigate the abnormalities in water and sodium metabolism, as well as the expression levels of arginine vasopressin receptor 1a (AVPR1a) and arginine vasopressin receptor 2 (AVPR2), in a rat model of chronic renal failure-chronic heart failure (CRF-CHF). One hundred male Sprague-Dawley rats were randomly assigned into four groups: the CG group (normal feeding), the CRF group (3/4 nephrectomy using a "two-step surgical method"), the CHF group (subcutaneous injection of isoproterenol at 100 mg/kg), and the CRF-CHF group (3/4 nephrectomy followed by a subcutaneous injection of isoproterenol at 100 mg/kg 1 week later). 4 weeks post-surgery, urine and blood samples were collected to measure 24 h urinary protein, sodium, and potassium levels. Serum creatinine (SCr) and blood urea nitrogen (BUN) levels were determined using assay kits. Left ventricular end diastolic pressure (LVEDP) and left ventricular systolic pressure (LVSP) were measured via left ventricular catheterization. The heart was weighed to calculate the left ventricular weight to body weight ratio (LVW/BW). The renal cortex and medulla were isolated to assess the relative mRNA and protein expression levels of AVPR1a and AVPR2. Compared to the CG group, the CRF and CRF-CHF groups exhibited significantly elevated levels of 24 h urinary protein, SCr, BUN, and relative expression levels of AVPR1a and AVPR2 in the renal cortex and medulla. The CHF and CRF-CHF groups showed significant increases in LVEDP and LVW/BW (P < 0.05). Additionally, compared to the CG group, the other three groups had significantly increased urinary sodium and blood potassium levels, and significantly decreased urinary potassium and blood sodium levels (P < 0.05). Compared to the CRF and CHF groups, the CRF-CHF group exhibited significantly higher levels of 24 h urinary protein, SCr, BUN, and relative expression levels of AVPR1a and AVPR2 in the renal cortex and medulla, along with significantly increased LVEDP and LVW/BW, significantly reduced LVSP, significantly increased urinary sodium and blood potassium levels, and significantly decreased urinary potassium and blood sodium levels (P < 0.05). Rats with CRF-CHF experienced exacerbated renal and cardiac failure, characterized by significant disturbances in water and sodium metabolism and abnormal expression of AVPR1a and AVPR2.
Collapse
Affiliation(s)
- Kun Sun
- Department of Nephropathy, Shandong Zibo Central Hospital, Zibo, 255036, Shandong, China
| | - Yingzhu Qiu
- Department of Spine Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200120, China
| | - Yao Sun
- Department of General Practice, Shandong ZiBo Central Hospital, Zibo, 255036, Shandong, China
| |
Collapse
|
10
|
Yang C, Diao L, Song Z, Guan C, Xu L, Bu Q, Jiang W, Yu H, Li D, Xu Y. Mesoporous activated carbon derived from Chinese herbal medicine residues for hemoperfusion removal of uremia toxins from progressive chronic kidney diseases patients. Heliyon 2024; 10:e38892. [PMID: 39498034 PMCID: PMC11533571 DOI: 10.1016/j.heliyon.2024.e38892] [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: 01/13/2024] [Revised: 09/03/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
Hemoperfusion is one of the most important therapies for progressive chronic kidney disease (CKD) and is effective at removing toxins from the blood. Increasing the efficiency of adsorbents applied in hemoperfusion is crucial. In the present study, shell of areca nut, one of the most common waste Chinese herb medicine residue with a porous structure was carbonized and activated at different temperatures to obtain two kinds of porous materials. The biocompatibility of the as-prepared porous materials was estimated via a hemolytic test, and the removal efficiency of the materials toward toxins was tested via an adsorption experiment in solution and blood samples from CKD patients, simulated hemoperfusion and in vivo hemoperfusion. After 4 h of adsorption, free and protein-bound toxins in solution were efficiently removed by the prepared porous materials, and the removal efficiency was better than that of commonly used hemoperfutor adsorbents. Most of the tested toxins can be removed from CKD blood samples and simulated hemoperfusion samples. Blood uremic toxins from CKD mice were also efficiently and safely removed after in vivo hemoperfusion using the as-prepared adsorbent. This work highlights promising adsorbents for hemoperfusion that could increase the therapeutic efficacy in patients with progressive CKD.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Lipeng Diao
- State Key Laboratory of Bio-fibers and Eco-textiles, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, PR China
| | - Zhuo Song
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Chen Guan
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Lingyu Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Quandong Bu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Huiqing Yu
- School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| | - Daohao Li
- State Key Laboratory of Bio-fibers and Eco-textiles, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, PR China
| | - Yan Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| |
Collapse
|
11
|
Liu Q, Pei Y, Xie Q, Bao W, Li X, Luan J, Han J. Renal Artery Coil Embolization as an Endovascular Approach for Establishing a Rabbit Model of Chronic Kidney Disease. J Vasc Interv Radiol 2024; 35:1234-1241.e3. [PMID: 38663515 DOI: 10.1016/j.jvir.2024.04.008] [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: 10/20/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/23/2024] Open
Abstract
PURPOSE To investigate the safety and feasibility of renal artery coil embolization for establishing chronic kidney disease (CKD) in rabbits. MATERIALS AND METHODS Ten male adult New Zealand rabbits underwent renal artery coil embolization. Initially, the main renal artery on 1 side was completely embolized, followed by embolization of approximately two-thirds of the primary branches of the contralateral renal artery 1 week later. Four rabbits were randomly chosen for sacrifice at 4 weeks after embolization, whereas the remaining 6 were sacrificed at 8 weeks after embolization. The assessment encompassed the animals' general condition, angiography, biochemical parameters, inflammatory markers, and histopathological examination of the kidneys and hearts. RESULTS Four weeks after embolization, serum creatinine level showed a substantial increase (2.4 mg/dL [SD ± 0.6]; P = .009 vs baseline), with a subsequent 4.12-fold elevation at 8 weeks after embolization (4.9 mg/dL [SD ± 1.4]; P < .001 vs baseline). Additionally, considerable increases in serum blood urea nitrogen, calcium, and potassium ions were observed at 8 weeks after embolization (58.3 mg/dL [SD ± 19.0]; P < .001 vs baseline; 23.1 mg/dL [SD ± 4.4]; P < .001 vs baseline; and 6.3 mEq/L [SD ± 0.7]; P < .001 vs baseline, respectively). The completely embolized kidney exhibited notable atrophy, severe fibrosis, and cortical calcification, whereas the contralateral partially embolized kidney displayed compensatory hypertrophy, along with glomerulosclerosis, tubular dilation, tubular casts, and interstitial fibrosis. CONCLUSIONS Renal artery coil embolization proved to be effective and safe for establishing a CKD model in rabbits.
Collapse
Affiliation(s)
- Qijia Liu
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Yun Pei
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Qian Xie
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Wenhan Bao
- Department of Nephrology, Peking University Third Hospital, Beijing, China
| | - Xuan Li
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jingyuan Luan
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jintao Han
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
12
|
Jiang Q, Yang Q, Zhang C, Hou C, Hong W, Du M, Shan X, Li X, Zhou D, Wen D, Xiong Y, Yang K, Lin Z, Song J, Mo Z, Feng H, Xing Y, Fu X, Liu C, Peng F, Wu L, Li B, Lu W, Yuan JXJ, Wang J, Chen Y. Nephrectomy and high-salt diet inducing pulmonary hypertension and kidney damage by increasing Ang II concentration in rats. Respir Res 2024; 25:288. [PMID: 39080603 PMCID: PMC11290206 DOI: 10.1186/s12931-024-02916-w] [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: 02/07/2024] [Accepted: 07/14/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a significant risk factor for pulmonary hypertension (PH), a complication that adversely affects patient prognosis. However, the mechanisms underlying this association remain poorly understood. A major obstacle to progress in this field is the lack of a reliable animal model replicating CKD-PH. METHODS This study aimed to establish a stable rat model of CKD-PH. We employed a combined approach, inducing CKD through a 5/6 nephrectomy and concurrently exposing the rats to a high-salt diet. The model's hemodynamics were evaluated dynamically, alongside a comprehensive assessment of pathological changes in multiple organs. Lung tissues and serum samples were collected from the CKD-PH rats to analyze the expression of angiotensin-converting enzyme 2 (ACE2), evaluate the activity of key vascular components within the renin-angiotensin-aldosterone system (RAAS), and characterize alterations in the serum metabolic profile. RESULTS At 14 weeks post-surgery, the CKD-PH rats displayed significant changes in hemodynamic parameters indicative of pulmonary arterial hypertension. Additionally, right ventricular hypertrophy was observed. Notably, no evidence of pulmonary vascular remodeling was found. Further analysis revealed RAAS dysregulation and downregulated ACE2 expression within the pulmonary vascular endothelium of CKD-PH rats. Moreover, the serum metabolic profile of these animals differed markedly from the sham surgery group. CONCLUSIONS Our findings suggest that the development of pulmonary arterial hypertension in CKD-PH rats is likely a consequence of a combined effect: RAAS dysregulation, decreased ACE2 expression in pulmonary vascular endothelial cells, and metabolic disturbances.
Collapse
Grants
- 82370063, 82170069, 82241012, 82120108001, 81970057, 82170065, 82000045, 82270052 National Natural Science Foundation of China
- 82370063, 82170069, 82241012, 82120108001, 81970057, 82170065, 82000045, 82270052 National Natural Science Foundation of China
- National Key Research and Development Program of China
Collapse
Affiliation(s)
- Qian Jiang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Qifeng Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chi Hou
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wei Hong
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Min Du
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xuanyi Li
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Dansha Zhou
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Dongmei Wen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Yuanhui Xiong
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Ziying Lin
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Jingjing Song
- Department of Stomatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Zhanjie Mo
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Yue Xing
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Xin Fu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Chunli Liu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Fang Peng
- Department of Critical Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Liling Wu
- Department of Nephrology, Shenzhen Second People's Hospital, Shenzhen, 518000, Guangdong, China
| | - Bing Li
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510320, Guangdong, China.
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
13
|
Liao X, Lu J, Huang Z, Lin J, Zhang M, Chen H, Lin X, Gao X, Gong S. Aminophylline suppresses chronic renal failure progression by activating SIRT1/AMPK/mTOR-dependent autophagy. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1311-1322. [PMID: 38808395 PMCID: PMC11532209 DOI: 10.3724/abbs.2024049] [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: 08/28/2023] [Accepted: 02/20/2024] [Indexed: 05/30/2024] Open
Abstract
Chronic renal failure (CRF) is a severe syndrome affecting the urinary system for which there are no effective therapeutics. In this study, we investigate the effects and mechanisms of aminophylline in preventing CRF development. A rat model of chronic renal failure is established by 5/6 nephrectomy. The levels of serum creatinine (SCR), urinary protein (UPR), and blood urea nitrogen (BUN) are detected by ELISA. Histological evaluations of renal tissues are performed by H&E, Masson staining, and PAS staining. Functional protein expression is detected by western blot analysis or immunofluorescence microscopy. Glomerular cell apoptosis is determined using the TUNEL method. Results show that Aminophylline significantly reduces the levels of SCR, UPR, and BUN in the CRF model rats. Histological analyses show that aminophylline effectively alleviates renal tissue injuries in CRF rats. The protein expression levels of nephrin, podocin, SIRT1, p-AMPK, and p-ULK1 are greatly increased, while p-mTOR protein expression is markedly decreased by aminophylline treatment. Additionally, the protein level of LC3B in CRF rats is significantly increased by aminophylline. Moreover, aminophylline alleviates apoptosis in the glomerular tissues of CRF rats. Furthermore, resveratrol promotes SIRT1, p-AMPK, and p-ULK1 protein expressions and reduces p-mTOR and LC3B protein expressions in CRF rats. Selisistat (a SIRT1 inhibitor) mitigates the changes in SIRT1, p-AMPK, p-ULK1, p-mTOR, and LC3B expressions induced by aminophylline. Finally, RAPA alleviates renal injury and apoptosis in CRF rats, and 3-MA eliminates the aminophylline-induced inhibition of renal injury and apoptosis in CRF rats. Aminophylline suppresses chronic renal failure progression by modulating the SIRT1/AMPK/mTOR-mediated autophagy process.
Collapse
Affiliation(s)
- Xin Liao
- Pediatric Departmentthe First Affiliated Hospital of Jinan UniversityGuangzhou510030China
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Jieyi Lu
- Pediatric Departmentthe First Affiliated Hospital of Jinan UniversityGuangzhou510030China
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Zhifeng Huang
- Department of Burns and Wound Repair SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Jinai Lin
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Miao Zhang
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Huanru Chen
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Xiaoqing Lin
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Xia Gao
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Sitang Gong
- Department of NephrologyGuangzhou Women and Children’s Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| |
Collapse
|
14
|
Zeper LW, Bos C, Leermakers PA, Franssen GM, Raavé R, Hoenderop JGJ, de Baaij JHF. Liver and spleen predominantly mediate calciprotein particle clearance in a rat model of chronic kidney disease. Am J Physiol Renal Physiol 2024; 326:F622-F634. [PMID: 38420675 DOI: 10.1152/ajprenal.00239.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Calciprotein particles (CPPs) provide an efficient mineral buffering system to prevent the complexation of phosphate and calcium in the circulation. However, in chronic kidney disease (CKD), the phosphate load exceeds the mineral buffering capacity, resulting in the formation of crystalline CPP2 particles. CPP2 have been associated with cardiovascular events and mortality. Moreover, CPP2 have been demonstrated to induce calcification in vitro. In this study, we examined the fate of CPP2 in a rat model of CKD. Calcification was induced in Sprague-Dawley rats by 5/6 nephrectomy (5/6-Nx) combined with a high-phosphate diet. Control rats received sham surgery and high-phosphate diet. Twelve weeks after surgery, kidney failure was significantly induced in 5/6-Nx rats as determined by enhanced creatinine and urea plasma levels and abnormal kidney histological architecture. Subsequently, radioactive and fluorescent (FITC)-labeled CPP2 ([89Zr]Zr-CPP2-FITC) were injected intravenously to determine clearance in vivo. Using positron emission tomography scans and radioactive biodistribution measurements, it was demonstrated that [89Zr]Zr-CPP2-FITC are mainly present in the liver and spleen in both 5/6-Nx and sham rats. Immunohistochemistry showed that [89Zr]Zr-CPP2-FITC are predominantly taken up by Kupffer cells and macrophages. However, [89Zr]Zr-CPP2-FITC could also be detected in hepatocytes. In the different parts of the aorta and in the blood, low values of [89Zr]Zr-CPP2-FITC were detectable, independent of the presence of calcification. CPP2 are cleared rapidly from the circulation by the liver and spleen in a rat model of CKD. In the liver, Kupffer cells, macrophages, and hepatocytes contribute to CPP2 clearance.NEW & NOTEWORTHY Calciprotein particles (CPPs) buffer calcium and phosphate in the blood to prevent formation of crystals. In CKD, increased phosphate levels may exceed the buffering capacity of CPPs, resulting in crystalline CPPs that induce calcification. This study demonstrates that labeled CPPs are predominantly cleared from the circulation in the liver by Kupffer cells, macrophages, and hepatocytes. Our results suggest that targeting liver CPP clearance may reduce the burden of crystalline CPP in the development of vascular calcification.
Collapse
Affiliation(s)
- Lara W Zeper
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter A Leermakers
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René Raavé
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
15
|
Dinh H, Kovács ZZA, Kis M, Kupecz K, Sejben A, Szűcs G, Márványkövi F, Siska A, Freiwan M, Pósa SP, Galla Z, Ibos KE, Bodnár É, Lauber GY, Goncalves AIA, Acar E, Kriston A, Kovács F, Horváth P, Bozsó Z, Tóth G, Földesi I, Monostori P, Cserni G, Podesser BK, Lehoczki A, Pokreisz P, Kiss A, Dux L, Csabafi K, Sárközy M. Role of the kisspeptin-KISS1R axis in the pathogenesis of chronic kidney disease and uremic cardiomyopathy. GeroScience 2024; 46:2463-2488. [PMID: 37987885 PMCID: PMC10828495 DOI: 10.1007/s11357-023-01017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
The prevalence of chronic kidney disease (CKD) is increasing globally, especially in elderly patients. Uremic cardiomyopathy is a common cardiovascular complication of CKD, characterized by left ventricular hypertrophy (LVH), diastolic dysfunction, and fibrosis. Kisspeptins and their receptor, KISS1R, exert a pivotal influence on kidney pathophysiology and modulate age-related pathologies across various organ systems. KISS1R agonists, including kisspeptin-13 (KP-13), hold promise as novel therapeutic agents within age-related biological processes and kidney-related disorders. Our investigation aimed to elucidate the impact of KP-13 on the trajectory of CKD and uremic cardiomyopathy. Male Wistar rats (300-350 g) were randomized into four groups: (I) sham-operated, (II) 5/6 nephrectomy-induced CKD, (III) CKD subjected to a low dose of KP-13 (intraperitoneal 13 µg/day), and (IV) CKD treated with a higher KP-13 dose (intraperitoneal 26 µg/day). Treatments were administered daily from week 3 for 10 days. After 13 weeks, KP-13 increased systemic blood pressure, accentuating diastolic dysfunction's echocardiographic indicators and intensifying CKD-associated markers such as serum urea levels, glomerular hypertrophy, and tubular dilation. Notably, KP-13 did not exacerbate circulatory uremic toxin levels, renal inflammation, or fibrosis markers. In contrast, the higher KP-13 dose correlated with reduced posterior and anterior wall thickness, coupled with diminished cardiomyocyte cross-sectional areas and concurrent elevation of inflammatory (Il6, Tnf), fibrosis (Col1), and apoptosis markers (Bax/Bcl2) relative to the CKD group. In summary, KP-13's influence on CKD and uremic cardiomyopathy encompassed heightened blood pressure and potentially activated inflammatory and apoptotic pathways in the left ventricle.
Collapse
Affiliation(s)
- Hoa Dinh
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Biochemistry, Bach Mai Hospital, Hanoi, 100000, Vietnam
| | - Zsuzsanna Z A Kovács
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Merse Kis
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Klaudia Kupecz
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Anita Sejben
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Gergő Szűcs
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Fanni Márványkövi
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Marah Freiwan
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Szonja Polett Pósa
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Katalin Eszter Ibos
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Éva Bodnár
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ana Isabel Antunes Goncalves
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, 6726, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Zsolt Bozsó
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gábor Tóth
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Andrea Lehoczki
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - László Dux
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
| | - Krisztina Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary
| | - Márta Sárközy
- Department of Biochemistry and Interdisciplinary Centre of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, 6720, Szeged, Hungary.
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6720, Hungary.
| |
Collapse
|
16
|
Mannon EC, Muller PR, Sun J, Bush WB, Coleman A, Ocasio H, Polichnowski AJ, Brands MW, O'Connor PM. NaHCO3 loading causes increased arterial pressure and kidney damage in rats with chronic kidney disease. Clin Sci (Lond) 2024; 138:189-203. [PMID: 38300615 DOI: 10.1042/cs20231709] [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: 12/20/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/02/2024]
Abstract
Sodium bicarbonate (NaHCO3) is commonly utilized as a therapeutic to treat metabolic acidosis in people with chronic kidney disease (CKD). While increased dietary sodium chloride (NaCl) is known to promote volume retention and increase blood pressure, the effects of NaHCO3 loading on blood pressure and volume retention in CKD remain unclear. In the present study, we compared the effects of NaCl and NaHCO3 loading on volume retention, blood pressure, and kidney injury in both 2/3 and 5/6 nephrectomy remnant kidney rats, a well-established rodent model of CKD. We tested the hypothesis that NaCl loading promotes greater volume retention and increases in blood pressure than equimolar NaHCO3. Blood pressure was measured 24 h daily using radio telemetry. NaCl and NaHCO3 were administered in drinking water ad libitum or infused via indwelling catheters. Rats were housed in metabolic cages to determine volume retention. Our data indicate that both NaHCO3 and NaCl promote hypertension and volume retention in remnant kidney rats, with salt-sensitivity increasing with greater renal mass reduction. Importantly, while NaHCO3 intake was less pro-hypertensive than equimolar NaCl intake, NaHCO3 was not benign. NaHCO3 loading significantly elevated blood pressure and promoted volume retention in rats with CKD when compared with control rats receiving tap water. Our findings provide important insight into the effects of sodium loading with NaHCO3 in CKD and indicate that NaHCO3 loading in patients with CKD is unlikely to be benign.
Collapse
Affiliation(s)
- Elinor C Mannon
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - P Robinson Muller
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Jingping Sun
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Weston B Bush
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Alex Coleman
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Hiram Ocasio
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Aaron J Polichnowski
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, U.S.A
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| |
Collapse
|
17
|
Li J, Nie J, Zhou Z, Guo M, Yang Q, Yuan D, Huang J, Li R, Li Q. Changes of FGF23 and hearing in chronic renal failure and their correlation analysis. Cytokine 2024; 174:156478. [PMID: 38134554 DOI: 10.1016/j.cyto.2023.156478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/01/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND To explore the association between fibroblast growth factor 23 (FGF23) and hearing in chronic renal failure (CRF). METHODS Pure tone audiometry was used to detect the hearing of patients with CRF; the level of serum FGF23, creatinine, blood urea nitrogen (BUN), parathyroid hormone (PTH), and mean binaural hearing threshold were compared to the control group (people without kidney disease). The rat model of renal failure was established by 5/6 nephrectomy, and the auditory brainstem response (ABR) of rats after modeling was detected by the Tucker Davis Technologies (TDT) system; the expression level of FGF23 in the peripheral blood, renal and cochlear tissue was also detected. RESULTS The incidence of hearing loss (HL) and serum FGF23 were higher in CRF patients than the control group; the sFGF23 was positively correlated with the mean binaural hearing threshold. Animal studies showed that the ABR threshold, creatinine, FGF23, BUN, and PTH increased after modeling; although, an increase in FGF23 was observed earlier than other indicators. The HL of rats with renal failure was significantly correlated with BUN, phosphate, PTH, sFGF23, kFGF23/β-actin, eFGF23/β-actin, weight, and modeling cycle. CONCLUSIONS Both CRF patients and rat models showed high-frequency HL. FGF23 was highly expressed in the serum of HL renal failure patients and rats, as well as in the renal tissue and cochlea of renal failure rats. Therefore, FGF23 may be involved in the occurrence and development of HL caused by CRF.
Collapse
Affiliation(s)
- Jiaqing Li
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jingwen Nie
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhu Zhou
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Min Guo
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Yang
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dunlu Yuan
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jingjing Huang
- Department of Medical Record, The Third People's Hospital of Kunming, Kunming, China
| | - Ruomei Li
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Li
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
18
|
Liang J, Liu Y. Animal Models of Kidney Disease: Challenges and Perspectives. KIDNEY360 2023; 4:1479-1493. [PMID: 37526653 PMCID: PMC10617803 DOI: 10.34067/kid.0000000000000227] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Kidney disease is highly prevalent and affects approximately 850 million people worldwide. It is also associated with high morbidity and mortality, and current therapies are incurable and often ineffective. Animal models are indispensable for understanding the pathophysiology of various kidney diseases and for preclinically testing novel remedies. In the last two decades, rodents continue to be the most used models for imitating human kidney diseases, largely because of the increasing availability of many unique genetically modified mice. Despite many limitations and pitfalls, animal models play an essential and irreplaceable role in gaining novel insights into the mechanisms, pathologies, and therapeutic targets of kidney disease. In this review, we highlight commonly used animal models of kidney diseases by focusing on experimental AKI, CKD, and diabetic kidney disease. We briefly summarize the pathological characteristics, advantages, and drawbacks of some widely used models. Emerging animal models such as mini pig, salamander, zebrafish, and drosophila, as well as human-derived kidney organoids and kidney-on-a-chip are also discussed. Undoubtedly, careful selection and utilization of appropriate animal models is of vital importance in deciphering the mechanisms underlying nephropathies and evaluating the efficacy of new treatment options. Such studies will provide a solid foundation for future diagnosis, prevention, and treatment of human kidney diseases.
Collapse
Affiliation(s)
- Jianqing Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
19
|
Sivertsson E, Ceder S, Nangaku M, Hansell P, Nordquist L, Palm F. AST-120 to Target Protein-Bound Uremic Toxins Improves Cardiac Output and Kidney Oxygenation in Experimental Chronic Kidney Disease. Kidney Blood Press Res 2023; 48:114-123. [PMID: 36791683 DOI: 10.1159/000529272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 01/11/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Chronic kidney disease (CKD) is a global health problem with increasing incidence which is closely associated with cardiac dysfunction. In CKD, uremic toxins accumulate as kidney function declines. Additionally, high salt intake is a growing health issue worldwide which can exacerbate kidney disease. In this study, we investigated the effect of reducing plasma levels of protein-bound uremic toxins in a rat model of CKD, challenged with high salt intake and compared the effects to those of conventional treatment using an angiotensin-converting enzyme inhibitor (ACEI). METHODS In rats, the right kidney and 2/3 of the left kidney were surgically removed (5/6 nephrectomy). Animals were fed a normal-salt diet and randomized to either no treatment (control) or chronic treatment with either the oral absorbent AST-120 to reduce plasma levels of protein-bound uremic toxins or the ACEI enalapril to inhibit angiotensin II signaling for 5 weeks. Following treatment, kidney function was measured before and after a week of high salt intake. Cardiac output and markers of oxidative stress were measured at the end of the study period. RESULTS Treatment with AST-120 resulted in decreased levels of the uremic toxin indoxyl sulfate, improved cardiac output (mL/min: AST-120 44.9 ± 5.4 compared to control 26.6 ± 2.0; p < 0.05), and decreased urinary oxidative stress. ACEI reduced oxidative stress in kidney tissue and improved the glomerular filtration rate in response to high salt intake (mL/min: ACEI 1.5 ± 0.1; compared to control 1.1 ± 0.1; p < 0.05). Both interventions improved intrarenal oxygen availability (mm Hg: AST-120 42.8 ± 0.8; ACEI 43.2 ± 1.9; compared to control 33.4 ± 1.3; p < 0.05). CONCLUSION AST-120 administered to reduce plasma levels of uremic toxins, such as indoxyl sulfate, has potential beneficial effects on both cardiac and kidney function. Targeting uremic toxins and angiotensin II signaling simultaneously could be an efficient strategy to target both cardiac and kidney dysfunction in CKD, to further slow progression of disease in patients with CKD.
Collapse
Affiliation(s)
- Ebba Sivertsson
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sara Ceder
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo, Japan
| | - Peter Hansell
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Lina Nordquist
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|