Keywords: atrophy, cachexia, mitochondria, renal, uremia
Abstract
Preclinical animal models of chronic kidney disease (CKD) are critical to investigate the underlying mechanisms of disease and to evaluate the efficacy of novel therapeutics aimed to treat CKD-associated pathologies. The objective of the present study was to compare the adenine diet and 5/6 nephrectomy (Nx) CKD models in mice. Male and female 10-wk-old C57BL/6J mice (n = 5–9 mice/sex/group) were randomly allocated to CKD groups (0.2−0.15% adenine-supplemented diet or 5/6 Nx surgery) or the corresponding control groups (casein diet or sham surgery). Following the induction of CKD, the glomerular filtration rate was reduced to a similar level in both adenine and 5/6 Nx mice (adenine diet-fed male mice: 81.1 ± 41.9 µL/min vs. 5/6 Nx male mice: 160 ± 80.9 µL/min, P = 0.5875; adenine diet-fed female mice: 112.9 ± 32.4 µL/min vs. 5/6 Nx female mice: 107.0 ± 45.7 µL/min, P = 0.9995). Serum metabolomics analysis indicated that established uremic toxins were robustly elevated in both CKD models, although some differences were observed between CKD models (i.e., p-cresol sulfate). Dysregulated phosphate homeostasis was observed in the adenine model only, whereas Ca2+ homeostasis was disturbed in male mice with both CKD models. Compared with control mice, muscle mass and myofiber cross-sectional areas of the extensor digitorum longus and soleus muscles were ∼18−24% smaller in male CKD mice regardless of the model but were not different in female CKD mice (P > 0.05). Skeletal muscle mitochondrial respiratory function was significantly decreased (19−24%) in CKD mice in both models and sexes. These findings demonstrate that adenine diet and 5/6 Nx models of CKD have similar levels of renal dysfunction and skeletal myopathy. However, the adenine diet model demonstrated superior performance with regard to mortality (∼20−50% mortality for 5/6 Nx vs. 0% mortality for the adenine diet, P < 0.05 for both sexes) compared with the 5/6 Nx surgical model.
NEW & NOTEWORTHY Numerous preclinical models of chronic kidney disease have been used to evaluate skeletal muscle pathology; however, direct comparisons of popular models are not available. In this study, we compared adenine-induced nephropathy and 5/6 nephrectomy models. Both models produced equivalent levels of muscle atrophy and mitochondrial impairment, but the adenine model exhibited lower mortality rates, higher consistency in uremic toxin levels, and dysregulated phosphate homeostasis compared with the 5/6 nephrectomy model.
INTRODUCTION
Chronic kidney disease (CKD) increases in prevalence with age, particularly in individuals with hypertension and diabetes (1). More than 35 million people in the United States are estimated to have CKD, which has no cure, and available treatment options remain limited to peritoneal dialysis, hemodialysis, and kidney transplantation (1, 2). CKD is a catabolic disease that results in loss of muscle mass and function that has been strongly associated with mortality (3). It has been established that muscle catabolism, but not malnutrition, is primarily responsible for this increased mortality risk in CKD (4). In addition, several uremic toxins are known to cause skeletal muscle atrophy through a variety of proposed mechanisms including activation of proteolytic and autophagy pathways (5–7), as well as mitochondrial dysfunction and oxidative stress (8–13). In this regard, recent studies have shown that skeletal muscle abnormalities including muscle wasting and mitochondrial dysfunction are prevalent in patients with renal failure and that uremic toxins may play a significant role in the developing skeletal myopathy (14–16).
Numerous preclinical models of CKD have been developed to investigate the molecular mechanisms underlying CKD pathobiology as well as to test new therapeutic approaches aimed to mitigate CKD-associated pathologies (14, 17, 18). The 5/6 nephrectomy (Nx) model, which involves surgical resection of kidney mass, is one of the most widely used techniques to successfully induce renal failure in animals (19, 20). However, complications such as the ureteral injury and bleeding contribute to a considerable mortality rate (up to 50%) (20). In addition, added experimental variance of this model due to slight differences in the size of the remnant kidney can significantly impact experimental outcomes.
Notably, the adenine diet model of CKD, a nonsurgical option, was originally developed to induce renal dysfunction in rats (21–24). Kidney pathology with adenine feeding stems from the formation of 2,8-dihydroxyadenine, an adenine metabolite that crystalizes within renal tubules and causes injury, inflammation, tubular atrophy, and fibrosis of the renal parenchyma (25). More recently, the adenine model of nephropathy has been successfully used in mice and provides better control over the disease severity with modification of adenine dosage (26, 27). For example, recent studies have reported that mice fed an adenine diet for 6 wk showed decreased kidney size and glomerular filtration rate (GFR), increased blood urea nitrogen (BUN), and renal fibrosis compared with mice fed a control diet (14, 28). Despite the growth in popularity of the adenine diet CKD model, no studies have directly compared the associated skeletal myopathy in the adenine model with the traditional surgical CKD models. Therefore, the primary goal of this study was to compare skeletal myopathy between the adenine diet and 5/6 Nx CKD models in both male and female mice. It was hypothesized that both CKD models would produce similar impairments in renal function and skeletal myopathy.
METHODS
Animals
Male and female 10-wk-old C57BL/6J mice (n = 5–9 mice/group/sex) were obtained from The Jackson Laboratory (Bar Harbor, ME). All rodents were housed in a light-controlled (12:12-h light-dark cycle), temperature-controlled (22°C), and humidity-controlled (50%) room, and maintained on standard chow (Teklad 2018, Envigo, 18.6% crude protein) before CKD induction with free access to food and water. All animal experiments adhered to the National Institutes of Health Guide for the Care and Use of Laboratory Animals and any updates. All procedures were approved by the Institutional Animal Care and Use Committee of the University of Florida and Malcom Randall Veterans Affairs Medical Center.
Adenine Diet-Induced Nephropathy
We used an established adenine diet model (17, 27, 29–32) to induce kidney disease in mice. Mice were allocated to a casein-based chow diet (20% casein) for 7 days, followed by the induction of renal tubular injury by supplementing the casein-based chow with 0.2% adenine diet for 7 days, followed by a maintenance diet supplemented with 0.15% adenine diet for 7 wk. CKD mice were then placed back on the control casein diet for 2 wk before the terminal experiments. Control animals received casein-based chow for the entirety of the study (Fig. 1A).
Figure 1.
Comparison of adenine nephropathy and 5/6 nephrectomy (Nx) models. A: schematic overview of the study design for adenine-induced nephropathy and 5/6 Nx. Male and female C57BL/6J mice were allocated to control (casein diet and sham surgery) and chronic kidney disease (CKD; adenine diet and 5/6 Nx) groups. Glomerular filtration rate (GFR) and blood urea nitrogen (BUN) were assessed 4–7 days prior to euthanization. B: probability of survival throughout the study duration. C: body weights in control and CKD mice. D: GFR measured by FITC-inulin clearance. E: BUN measured using a commercial kit. GFR and BUN tests were conducted 8 wk after adenine diet in the group of diet-induced nephropathy and 4 wk after the second-step surgery in the group of 5/6 Nx. Values are represented as means ± SD; n = 4–9 mice/group/sex. Data were analyzed using two-way ANOVA, and Tukey’s post hoc analysis was performed when appropriate. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 vs. control; ###P < 0.001 vs. adenine. FITC
5/6 Nx Model
A modified 5/6 Nx model (20) was used to surgically induce renal dysfunction in mice. This consisted of two surgical steps: 1) initial upper and lower pole ligation of the left kidney during the first surgery and 2) right kidney resection 1 wk after the first surgery (Fig. 1A). All operating procedures were performed under aseptic techniques. Mice were anesthetized with 1.0−1.2% isoflurane mixture (2% during the induction phase) and placed on a warm pad (SpaceGel, Braintree Scientific) during the surgery. In the supine position, a midline abdominal incision was made, and the left kidney was exposed. The surgeon performed careful separation of perirenal fat, connective tissue, the adrenal gland, and the ureter with blunt forceps. A 3-0 silk suture was placed circumferentially around the upper and lower poles of the left kidney. Thereafter, application of the ligatures was performed, and the renal parenchyma was observed to ensure the presence of ischemia without bleeding. Subsequently, the contralateral right renal Nx was performed 7 days after the first operation. The Nx was performed by reopening the previous midline laparotomy incision, and the right kidney was isolated. Care was taken to preserve the adrenal gland, after which the renal pedicle with the artery, vein, and ureter was ligated. Finally, the right kidney was extirpated by transecting the vessels and ureter immediately distal to the ligature. The abdominal muscle layer and skin were closed with 5-0 sterile absorbable suture using a simple continuous technique. Following each surgery, mice were treated with buprenorphine (0.1 mg/kg body weight). The sham surgery group also underwent two-step surgeries but without pole ligation of the left kidney or right kidney resection.
Assessment of Kidney Function
Kidney function was assessed by measuring GFR via FITC-inulin clearance as previously described (14, 33, 34). Briefly, FITC-inulin was dissolved in 0.9% NaCl [5% (wt/vol)], and the solution was dialyzed with a 1,000-kDa dialysis membrane (Spectrum Laboratories) in 0.9% NaCl at room temperature in the dark for 24 h, followed by sterile filtering through a 0.22-µm filter (Thermo Fisher). Under brief isoflurane anesthesia, FITC-inulin (2 µL/g body weight) was injected into the retroorbital sinus. The mouse was then returned to a clean cage, and ∼20 µL of blood was collected in heparinized capillary tubes via a ∼1-mm tail snip in the conscious mouse at 3, 5, 7, 10, 15, 35, 56, and 75 min following the injection. Thereafter, blood was centrifuged for 10 min at 4,000 rpm (4°C), and the plasma supernatant was transferred to a new tube. To measure the FITC-inulin concentration, plasma was diluted (1:20) in 0.5 mol/L of HEPES buffer (pH 7.4) and loaded into a 96-well plate along with a FITC-inulin standard curve, and fluorescence was detected using a BioTek Synergy II plate reader. GFR was calculated using a two-phase exponential decay curve fit in GraphPad Prism as previously described (14, 33, 34), and was not normalized to body weight. BUN was also assessed using a commercial kit (K024, Arbor Assays) according to the manufacturer’s instructions.
Serum Biochemical Analyses
Serum levels of calcium and inorganic phosphate were determined by a spectrophotometer using commercial kits (Cat. No. P7516 and C7529, Pointe Scientific) according to the manufacturer’s instructions. Quantification of serum parathyroid hormone (PTH) and fibroblast growth factor-23 (FGF23) was performed using ELISA (Cat. No. 60–2305 and 60–6800, Immutopics/Quidel) according to the manufacturer’s instructions. Serum used in these analyses was collected at the time of euthanasia.
Serum Metabolomics
Analysis of serum metabolomics was carried out at the Southeast Center for Integrated Metabolomics at the University of Florida (http://secim.ufl.edu) as previously described (35). On the day of euthanization, whole blood was collected via cardiac puncture with a 21-gauge needle syringe, allowed to clot for ∼20 min at room temperature, and centrifuged at 4,000 rpm for 10 min. The supernatant was transferred to new Eppendorf tubes and stored at −80°C until analysis. Serum samples were processed using a metabolomics platform to quantify select tryptophan-derived metabolites that are established uremic toxins. Twenty-five microliters of serum from each mouse (n = 4 mice/sex/group) were mixed with 5 µL of internal standard solution containing [1³C11]tryptophan, serotonin D4, kynurenine D4, kynurenic Acid D5, xanthurenic acid D4, [1³C6]anthranilic acid, [1³C]indoxyl sulfate, p-cresol sulfate D7, and 3-indole-acetate D7. After the addition of 200 µL of acetonitrile:methanol:acetone (8:1:1) with 0.1% formic acid, samples were placed in an ultrasonic bath for 10 min and then centrifuged at 20,000 g for 5 min at 4°C to pellet the protein. Then, 190 µL of supernatant were transferred from each sample into clean tubes and dried under a gentle stream of nitrogen at 30°C. The dried extracts were resuspended with 25 µL water with 0.1% formic acid and incubated at 4°C for 10−15 min, and the samples were then centrifuged at 20,000 g for 5 min at 4°C. Supernatants were transferred into clean LC vials for targeted LC-MS quantitation on a Thermo Q-Exactive Oribtrap mass spectrometer with Dionex UHPLC and autosampler. Tryptophan, serotonin, kynurenine, kynurenic acid, xanthurenic acid, and anthranilic acid were quantified in positive ionization, whereas indoxyl sulfate, p-cresol sulfate, and 3-indole-acetate were quantified in negative ionization. Separation was achieved on an ACE C18-PFP (C18 bonded HPLC column with a pentafluorophenyl phase) 100 × 2.1-mm, 2-µm column, using a gradient with mobile phase A as 0.1% formic acid in water and mobile phase B as acetonitrile. The flow rate was 350 µL/min with a column temperature of 25°C and injection volume of 2 µL. A nine-point calibration curve and quality control samples were prepared for targeted quantitation of tryptophan, serotonin, kynurenine, kynurenic acid, xanthurenic acid, anthranilic acid, indoxyl sulfate, p-cresol sulfate, and 3-indole-acetate. Twenty microliters of each calibrator and quality controls were supplemented with 5 µL of indoxyl sulfate. Peak areas of each analyte and the corresponding internal standards in the calibrator, quality controls, and samples were integrated using Xcalibur 4.0. A calibration curve was generated by plotting nominal concentration of the analyte in the calibrators versus the peak area ratio of the analyte and internal standard. Quality controls and samples were quantitated against the calibration curve.
Immunofluorescence Microscopy
Skeletal muscle fiber cross-sectional area (CSA) was assessed by immunofluorescence microscopy. Isolated extensor digitorum longus (EDL) and soleus (SOL) muscles were mounted in disposable base molds (Cat. No. 6235215, Electron Microscopy Science) using optimum cutting temperature mounting medium (Tissue-Tek, Sakura Finetek) and frozen in liquid nitrogen-cooled isopentane for cryosectioning. Transverse serial sections (10 µm) were cut from the midbelly of the skeletal muscles using a Leica 3050S cryostat at −20°C, mounted on frosted microscope slides, briefly air-dried at room temperature, and stored at −80°C until being stained. Prior to staining, frozen muscle sections were air-dried at room temperature for 10 min and fixed with 4% paraformaldehyde for 5 min. Following 2 × 3-min washes with 1× PBS, sections were permeabilized with 0.3% Triton X-100 in PBS for 10 min at room temperature. After 3 × 5-min washes with PBS, sections were incubated in blocking buffer (5% goat serum and 1% BSA in PBS) for 1 h at room temperature. To label the sarcolemma, sections were incubated with primary antibody against to laminin (L9393, Sigma, 1:100) overnight at 4°C. Following 4 × 5-min washes with 1× PBS, muscle sections were stained with secondary antibody (Alexa Fluor 488 goat anti-rabbit IgG, 1:250) diluted in blocking buffer. Following 4 × 5-min washes with 1× PBS, coverslips were mounted with Vectashield hardmount (H-1500, Vector Laboratories). Slides were imaged at ×20 magnification with an Evos FL2 Auto microscope (Thermo Scientific), and tiled images of the entire muscles were obtained for analysis. Quantification of myofibers CSA was performed using MuscleJ (36), an automated analysis software developed in Fiji.
Skeletal Muscle Mitochondrial Isolation
Skeletal muscle mitochondria were isolated as previously described (16, 37). Briefly, the following skeletal muscles were dissected: tibialis anterior (TA), quadriceps, hamstrings, gastrocnemius, plantaris, triceps, pectorals, gluteus maximus, and erector spinae muscles and placed in ice-cold mitochondrial isolation medium (300 mM sucrose, 10 mM HEPES, and 1 mM EGTA, pH 7.1). Muscles were then trimmed to remove excessive blood, connective tissue, and fat, minced in a petri dish on ice, transferred to ice-cold mitochondrial isolation medium supplemented with BSA (1 mg/mL), homogenized on ice using a glass-Teflon homogenizer (Wheaton), and subsequently centrifuged at 800 g for 10 min to pellet nonmitochondrial myofibrillar proteins, nuclei, and other cellular components. The resulting supernatant was centrifuged at 10,000 g for 10 min to pellet mitochondria. All centrifugation steps were performed at 4°C. The mitochondrial pellet was gently washed to remove damaged mitochondria and then resuspended in mitochondrial isolation medium (without BSA). The protein concentration was determined using a bicinchoninic acid protein assay (SL256970, Thermo Scientific).
Measurement of Mitochondrial Respiratory Function
Mitochondrial respiratory function was assessed using high-resolution respirometry (Oroboros Oxygraph-2K) at 37°C in mitochondrial assay buffer (105 mM K-MES, 30 mM KCl, 1 mM EGTA, 10 mM K2HPO4, 5 mM MgCl2·6H2O, and 2.5 mg/mL BSA, pH 7.2) supplemented with 20 mM creatine. Twenty micrograms of mitochondria were added to the Oxygraph-2K chamber filled with 2 mL of mitochondrial assay buffer. To energize the mitochondria, 5 mM pyruvate and 2.5 mM malate substrates were provided as fuel (state 2), followed by the sequential additions of 4 mM ADP (complex I state 3), 0.005 mM cytochrome c (to check the integrity of the mitochondrial outer membrane), and 10 mM succinate (complex I + II state 3).
Statistical Analysis
All statistical analyses were conducted using GraphPad Prism software (v. 8.3.1). Data were analyzed using two-way ANOVA with Tukey’s post hoc testing for multiple comparisons when appropriate. The two-stage step-up method of Benjamini, Krieger, and Yekutieli was used for multiple comparisons of mitochondrial respiration data. When sex differences or group × sex interactions were detected, data were presented accordingly. Pearson correlations were performed using two-tailed statistical testing. In all cases, P < 0.05 was considered statistically significant. All data were presented as means with error bars representing SDs.
RESULTS
Characterization of Renal Function and Mortality in the Adenine Diet and 5/6 Nx CKD Models
The widely used 5/6 Nx model of CKD involves two invasive surgeries that produce a rapid decrease in kidney mass that does not mimic the progressive nature of CKD in humans. The mortality rate in surgical models of CKD complicates preclinical study design. To this end, there was a notable difference in the survival rate between adenine and 5/6 Nx CKD models in both male and female mice. A total of 10 male mice and 11 female mice underwent the 5/6 Nx surgery, and five male mice and two female mice died throughout the experiment. In contrast, no mortality was observed for male or female CKD mice subjected to the adenine diet (Fig. 1B). Average body weights of mice fed with the casein diet were not different from mice with sham surgery in both sexes. Both adenine feeding and 5/6 Nx led to reduced body weight in male mice only (Fig. 1C). GFR was decreased in both adenine and 5/6 Nx mice compared with the corresponding control mice (Fig. 1D). Importantly, no difference in GFR was found between adenine and 5/6 Nx CKD models (Fig. 1D). Similar to GFR, BUN was significantly higher in animals with CKD (Fig. 1E). Interestingly, BUN levels were significantly greater (although highly variable) in 5/6 Nx mice compared with adenine diet-fed mice (Fig. 1E).
Dysregulated mineral and bone metabolism is common in patients with CKD. To characterize biochemical markers related to mineral and bone metabolism, we compared serum levels of phosphate, Ca2+, FGF23, and PTH in both CKD models (Fig. 2). Baseline concentrations of these serum biochemical markers between casein diet-fed and sham controls were similar in both male and female mice (Fig. 2). Interestingly, serum phosphate (Fig. 2A) and FGF23 (Fig. 2B) levels were significantly increased in adenine diet-fed mice but not in 5/6 Nx mice. Serum Ca2+ levels were significantly increased in both CKD models for male but not female mice (Fig. 2C), indicating a clear sex difference. PTH was significantly greater in male CKD mice compared with controls; however, male adenine diet-fed mice displayed higher PTH compared with 5/6 Nx male mice (Fig. 2D). Taken together, these data provide support for the clinical relevance of the adenine model and highlight sex differences in preclinical CKD models that are not examined often.
Figure 2.
Serum biochemical markers associated with dysregulated bone and mineral metabolism. Serum phosphate (A), fibroblast growth factor-23 (FGF23; B), calcium (C), and parathyroid hormone (PTH; D) were determined using a microplate spectrophotometer. Values are represented as means ± SD; n = 3 or 4 mice/group/sex. Data were analyzed using a two-way ANOVA, and Tukey’s post hoc analysis was performed when appropriate. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 vs. control; ##P < 0.01 and ###P < 0.001 vs. adenine. Nx, nephrectomy.
Adenine Diet and 5/6 Nx Models Increase Uremic Toxins
With impaired renal function, there is often increased accumulation of circulating uremic toxins, which have been strongly associated with CKD pathobiology (38–43). For this reason, we performed targeted serum metabolomics analyses to examine the levels of uremic toxins in adenine and 5/6 Nx CKD models (Fig. 3). Consistent with previous reports (28, 44, 45), mice with adenine-induced and 5/6 Nx-induced CKD displayed increases in serum levels of established uremic toxins, including kynurenine (Fig. 3A), kynurenic acid (Fig. 3B), xanthurenic acid (Fig. 3C), and indoxyl sulfate (Fig. 3E). However, there were some differences between sexes and CKD models that were noteworthy. Interestingly, p-cresol sulfate, a well-known uremic toxin linked to cardiovascular disease risk and insulin resistance (41, 46), was elevated only in adenine diet-fed mice (Fig. 3F). Similarly, indole-3-acetate was elevated in male adenine diet-fed mice (but not female mice), whereas 5/6 Nx male mice displayed no differences from sham controls (Fig. 3G). There were no changes in the concentrations of serum tryptophan (Fig. 3H) and serotonin (Fig. 3I) in male and female mice in either CKD model. Another striking observation was that the variability in uremic toxin levels was substantially greater in 5/6 Nx groups, suggesting that the adenine diet model produces more consistent levels of uremic toxins.
Figure 3.
Serum uremic toxins in adenine nephropathy and 5/6 nephrectomy (Nx) models. Serum metabolomics determined circulating uremic toxins derived from tryptophan metabolism measured via mass spectrometry (LC-MS/MS). A: kynurenine. B: kynurenic acid. C: xanthurenic acid. D: anthranilic acid. E: indoxyl sulfate. F: p-cresol sulfate. G: indole-3-acetate. H: tryptophan. I: serotonin. Values are represented as means ± SD; n = 4 mice/group/sex. Data were analyzed using two-way ANOVA, and Tukey’s post hoc analysis was performed when appropriate. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 vs. control; #P < 0.05, ##P < 0.01, ###P < 0.001, and ###P < 0.0001 vs. adenine.
Muscle Mass and Myofiber CSA Are Decreased in Male but Not Female CKD Mice
Muscle catabolism is a common feature of CKD contributing to the progressive myopathic symptoms including muscle atrophy, weakness, and impaired physical function. To compare the muscle pathology in these CKD models, we measured muscle mass and myofiber CSAs in fast-twitch EDL and slow-twitch SOL muscles. There were no differences in muscle mass for EDL and SOL muscles between the control groups (casein diet vs. sham surgery). Both adenine diet-fed and 5/6 Nx male CKD mice had significantly reduced muscle mass in both EDL and SOL muscles (Fig. 4, B and G). Interestingly, EDL and SOL muscle masses were not statistically different between CKD and control female mice (Fig. 4, B and G). Consistent with muscle masses, mean myofiber CSA was not different between casein diet-fed and sham-operated mice (Fig. 4, C and H). Adenine diet-fed and 5/6 Nx male mice displayed significantly smaller myofiber CSA in EDL and SOL muscles (Fig. 4, C and H). Similar to muscle mass, mean myofiber CSAs of EDL and SOL muscles were not different between CKD and control groups in female mice (Fig. 4, C and H).
Figure 4.
Chronic kidney disease causes muscle wasting and reduced fiber cross-sectional area (CSA) in male but not in female mice. A: representative immunofluorescence image of the extensor digitorum longus (EDL) muscle stained with laminin from a casein diet-fed control mouse. B: muscle mass for the EDL muscle. C: mean myofiber CSA of the EDL muscle. D and E: relative frequency distributions of EDL myofiber CSA for male mice (D) and female mice (E). F: representative immunofluorescence image of the soleus (SOL) muscle stained with laminin from a casein diet-fed control mouse. G: muscle mass for the SOL muscle. H: mean myofiber CSA of the SOL muscle. I and J: relative frequency distributions of SOL myofiber CSA for male mice (I) and female mice (J). Values are represented as means ± SD; n = 5 mice/group/sex. Data were analyzed using two-way ANOVA, and Tukey’s post hoc analysis were performed when appropriate. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 vs. control; #P < 0.05 vs. casein. Nx, nephrectomy.
Skeletal Muscle Mitochondrial Respiratory Function Is Equally Impaired in Adenine Diet-Fed and 5/6 Nx Mice
Accumulating evidence from human and animal studies suggests that skeletal muscle mitochondrial abnormalities contribute to the myopathic symptoms in CKD (14, 28). Thus, we assessed mitochondrial respiratory function to determine how both CKD models impact muscle mitochondrial health. Figure 5A shows an overview of mitochondrial respiratory steps and conditions used in this experiment. First, we energized mitochondria in the absence of adenylates with pyruvate and malate to measure state 2 respiration, which displayed a significant group (CKD) effect (Fig. 5B). Next, maximal state 3 respiration was measured following the addition of 4 mM ADP, which assesses the maximal respiratory capacity supported by substrates that provide electrons to complex I (Fig. 5C). Next, exogenous cytochrome c was added to assess the integrity of the outer mitochondrial membrane (Fig. 5D). Finally, the subsequent addition of succinate allows additional electrons to enter the electron transport system through complex II (succinate dehydrogenase), facilitating analysis of maximal state 3 respiration supported by complex I + II substrates (Fig. 5E), which displayed a significant group (CKD) effect, but no significant effects of sex or interactions were detected. Notably, both adenine diet-fed and 5/6 Nx mice exhibited a similar magnitude (∼20%) of reduction in respiratory function, confirming that both murine CKD models impact muscle mitochondria to a similar degree.
Figure 5.
Mitochondrial respiratory function in skeletal muscle. Skeletal muscle mitochondria were isolated from control and chronic kidney disease mice. A: graphical depiction of the detailed respiratory protocol used. B: state 2 respiration supported by pyruvate and malate. C: ADP-stimulated mitochondrial respiration supported by pyruvate and malate. D: state 3 respiration following the addition of cytochrome c (Cyt C; used for checking mitochondrial integrity). E: state 3 respiration supported by pyruvate, malate, and succinate. Values are represented as means ± SD; n = 5–9 mice/group/sex. Data were analyzed using two-way ANOVA and the two-stage step-up method of Benjamini, Krieger, and Yekutieli for multiple comparisons. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. control. Nx, nephrectomy.
Pearson Correlation Analysis across Outcome Measures
To examine potential relationships between outcome measures, we next performed Pearson correlation analyses within each sex and model (Fig. 6). The results of these analyses revealed strong associations among GFR, uremic toxin levels, muscle size, and mitochondrial respiratory capacity (P < 0.05; Fig. 6). Elevated uremic toxins, including kynurenic acid, had inverse relationships with muscle mass and myofiber CSA (P < 0.05). Notably, several uremic toxins derived from kynurenine and indole pathways exhibited inverse relationships with mitochondrial respiratory function consistent with previous work (16, 28).
Figure 6.
Pearson correlation analyses across outcome measures. Pearson correlation coefficients among glomerular filtration rate (GFR), serum metabolites, muscle mass, myofiber cross-sectional area (CSA), and mitochondrial respiratory capacity were performed according to model and sex. Heatmaps were created from the Pearson correlation matrixes for adenine-induced nephropathy in male (A) and female (C) mice and for 5/6 nephrectomized (Nx) male (B) and female (D) mice. n = 4–9 mice/group/sex. *Significant correlation (P < 0.05). AA, anthranilic acid; EDL, extensor digitorum longus; I-3-A, indole-3-acetic acid; IS, indoxyl sulfate; KA, kynurenic acid; p-CS, p-cresol sulfate; SOL, soleus; XA, xanthurenic acid.
DISCUSSION
In the present study, two murine models of CKD, adenine diet and 5/6 Nx, were compared in disease severity and in relation with their adverse impact on skeletal muscle physiology. Although both models produced equivalent levels of renal dysfunction and impaired mitochondrial respiratory capacity in both sexes and muscle atrophy in male mice, the 5/6 Nx model displayed higher mortality in both sexes compared with the adenine diet model. Both models resulted in increased levels of established uremic toxins, including kynurenine, kynurenic acid, and indoxyl sulfate. However, several uremic toxins (p-cresol sulfate and indole-3-acetate) were increased in the adenine diet model but not in the 5/6 Nx model. Moreover, 5/6 Nx mice displayed greater variability in uremic toxin levels compared with adenine diet-fed mice. Interestingly, the pathophysiological impact of CKD was greater in male mice than in female mice regardless of the CKD model. Finally, we observed strong correlations between kidney function and the level of uremic toxins with muscle atrophy and mitochondrial function.
To confirm renal dysfunction in both models, we measured GFR, a gold standard measure of kidney function (34), as well as the concentration of BUN. Several previous studies using adenine diet-induced nephropathy or 5/6 Nx have reported reduced GFR and high levels of circulating urea nitrogen compared with their respective control groups (14, 28, 47). As expected, in the present study, GFR was significantly decreased in both CKD models compared with control animals. Importantly, GFR was not different between adenine diet-fed and 5/6 Nx mice for either sex. Similarly, BUN was elevated after the induction of renal dysfunction in male adenine diet-fed and 5/6 Nx mice in both sexes. Despite similar GFRs, BUN was significantly higher in 5/6 Nx mice compared with adenine diet-fed mice. There are several possible explanations for this result. First, protein catabolism with kidney disease resulting in increased nitrogenous waste is the primary source of urea production. However, given the comparable changes in body weights and EDL and SOL muscle masses in the two CKD models, rates of muscle protein catabolism are likely similar between models. This presumption is supported by previous studies that have reported similar rates of muscle protein degradation in 5/6 Nx and adenine models (5, 28). Second, the amount of dietary protein intake is associated with elevated levels of BUN in the CKD condition (48). In this regard, the average daily food intake is less in adenine diet-fed mice due in part to their reluctance to consume adenine-supplemented food (26). In a current study, we estimated daily food consumption in male and female mice consuming the adenine diet to be 2.4 and 2.1 g/day, respectively (28). This food intake is lower than the standard rodent diets (3–4 g/day), which were provided to the 5/6 Nx mice. Consistent with this notion, previous reports in young male C57BL/6J mice subjected to subtotal Nx found food consumption of 3–4 g of standard chow with 17% crude protein (49, 50). Accordingly, it is likely that the greater BUN concentration in 5/6 Nx mice compared with adenine diet-fed mice related to greater dietary protein intake.
Various animal models have been developed to characterize the numerous clinical features of a patient with CKD. Although no model has yet to fully recapitulate the complex array of clinical pathophysiology of patients with CKD, animal models of CKD remain a crucial tool for discovery of disease mechanisms and therapeutic development. To better understand potential differences in the 5/6 Nx and adenine diet models, we assessed several factors related to phosphate and Ca2+ homeostasis that have been linked to vascular calcification and increased cardiovascular events (51), and dysregulated bone and mineral metabolism, which are desirable features of preclinical CKD studies (26, 52). In the present study, we observed that male and female mice exposed to adenine diet had increases in serum phosphate and FGF23 levels similar to the results of a previous study (26). In contrast, neither phosphate nor FGF23 levels were changed in the 5/6 Nx group in either male or female mice. These results provide clear evidence of bone and mineral dysregulation in adenine diet-fed male mice. Notably, serum Ca2+ and PTH levels were similar in control and CKD female mice, again highlighting the sex differences in preclinical CKD models. The underlying mechanisms for this protection in female mice is not known but suggests that young female mice with intact ovaries are resistant to Ca2+ and phosphate dysregulation in CKD models.
Although there was a similar degree of renal dysfunction observed in both models, it is important to note that there was no mortality in the adenine diet-fed group but that there was 20−50% mortality in the 5/6 Nx group, with a greater rate observed in male mice. This observation confirms previous reports (26–28, 53) that used adenine diet models with <0.2% adenine in mice. The lower survival rate in the 5/6 Nx groups may be due in part to kidney tissue damage stemming from forced ligation that may cause intrarenal bleeding, damage on the ureter, or possible infection during the operation and postoperative care (17, 20, 26, 27). With regard to the 5/6 Nx model, it is important to highlight that this study performed a pole ligation 5/6 Nx model, which has subtle differences, including lower mortality rates, compared with conventional electrocoagulation models that carry greater risk of bleeding and infection (54). Although these differences in surgical approach for the 5/6 Nx model should not be overlooked, both models have been shown to produce similar levels of renal dysfunction and histopathology (54). In addition to lower mortality rates, the adenine model circumvents the potential confounding impact of subtle surgical differences on renal and systemic pathophysiology. Considering the cost, time, technical ability, and effort to perform the surgery and postoperative care, as well as the increased variability in outcomes (i.e., GFR, BUN, FGF23, PTH, serum phosphate and Ca2+, and uremic toxins levels) reported herein, there is appreciable value in using the adenine diet model in preclinical research.
As a result of insufficient kidney function, patients with CKD display increased levels of uremic toxins, which have been linked to chronic inflammation, vascular calcification, and oxidative stress, contributing to the high risk of cardiovascular complications (55, 56). In addition, uremic toxins have been shown to negatively impact skeletal muscle health (15, 57, 58). For example, accumulation of serum uremic toxins in patients with kidney disease is associated with sarcopenia and mitochondriopathy (28, 39, 59). These pathological effects may be worsened in severe stages of renal disease (i.e., end-stage renal disease) despite treatment efforts such as hemodialysis because of incomplete filtration of uremic toxins by the conventional dialysis membrane, particularly for protein-bound toxins (60). In this study, we examined tryptophan-derived uremic toxins in both adenine and 5/6 Nx models of CKD. Both murine CKD models produced similar profiles of increased uremic toxins, including indoxyl sulfate and kynurenines, consistent with earlier studies (61, 62). Despite similar changes in kynurenines and indoxyl sulfate, there were noteworthy differences between models. For example, p-cresol sulfate, which plays a significant role in endothelial dysfunction and is associated with cardiovascular disease risk (41, 46), was only elevated in adenine diet-fed mice. The exact mechanisms underlying the difference of p-cresol sulfate levels between adenine and 5/6 Nx murine CKD models is not known at this time. As shown in Fig. 3, the adenine model produced more consistent increases in uremic toxins compared with 5/6 Nx mice. This is demonstrated by the standard deviations for key uremic toxins such as indoxyl sulfate, kynurenine, and kynurenic acid. Further analysis using Pearson correlations revealed that there were significant inverse correlations between GFR and uremic toxins in both models of CKD (Fig. 6).
Muscle wasting, defined as the loss of muscle mass and reduced fiber CSA, is one of the most notable characteristics observed in patients with CKD, and contributes to exercise intolerance, frailty, and poor quality of life (63, 64). In the CKD condition, negative net protein balance in which the rate of protein degradation is greater than the rate of protein synthesis has been known to contribute to the increased whole body protein turnover (7). Numerous cellular pathways have been shown to contribute to elevated protein catabolism in CKD, including impaired satellite cell function and muscle growth (65), suppressed anabolic cell signaling such as insulin/insulin-like growth factor-I-phospho-AKT-mammalian target of rapamycin-p70S6 kinase pathways (66), elevated protein degradation (i.e., caspase 3 and the ubiquitin-proteasome system) (67), increased myostatin (a negative regulator of muscle growth) (68), and altered microRNA expression (69). In addition to the factors such as inflammation and oxidative stress, uremic toxicity plays a significant role in the catabolic condition in CKD (39). The role of tryptophan-derived metabolites, such as kynurenine and indoxyl sulfate, in muscle wasting has been suggested (15, 57, 58). For example, Berru et al. (14) showed that C2C12 cells in DMEM growth medium supplemented with 5% serum acquired from adenine-induced CKD mice for 24 h resulted in myotube atrophy. McGee-Lawrence et al. (58) showed that treatment with kynurenine through intraperitoneal injection into young healthy mice (with normal kidney function) resulted in decreased muscle fiber size. Similarly, Sato et al. (15) reported that adenine diet-induced CKD mice accumulated indoxyl sulfate in muscle tissues and decreased myofiber CSA in the tibialis anterior muscle. In the present study, we observed that male CKD mice, regardless of model, displayed smaller muscle mass and myofiber CSA in the EDL and SOL muscles, which were strongly associated with elevated serum concentrations of uremic toxins (Fig. 6). The exact mechanisms by which uremic toxins may mediate muscle atrophy are not fully known but represents an exciting avenue for therapeutic discovery.
In contrast to male CKD mice, muscle mass and fiber CSA were modestly decreased (6–13%) in female CKD mice compared with their respective controls; however, this was not statistically significant. A plausible explanation of the sex-dependent impact of CKD on muscle wasting may be related to the protective effects of estrogen on skeletal muscle because the female mice used in this study were young (10 wk old) and had normal menstrual cycles: two characteristics that do not match female patients with CKD well. Clinical studies have reported that CKD is more prevalent in postmenopausal women (70), but also that menopause accelerates the disease progression, including skeletal muscle weakness (71). Accumulating evidence from animal studies indicates that estrogen has a protective role in the development of renal disease (72), bone metabolism (73), regulation of muscle mass (74, 75), and mitochondrial function (76, 77). Consistent with this hypothesis, estrogen deficiency induced by ovariectomy resulted in more severe glomeruloscelerosis and increased BUN compared with animals with intact ovaries (72). Kitajima et al. (78) reported that surgical removal of the ovaries from female mice results in muscle atrophy, which could be rescued by estradiol administration. Taken together, the results of the present study and others (28, 78) suggest the need to consider the age, menstrual status, and role of estrogens in preclinical CKD studies involving female mice.
In addition to muscle atrophy, skeletal muscle abnormalities prevalent in CKD include impaired mitochondrial biogenesis and/or mitochondrial function, which is believed to contribute to poor physical performance and exercise intolerance (9, 28, 79–81). Kestenbaum et al. (80) showed that compared with age-matched controls with normal renal function, patients with CKD had poor physical performance (6-min walk test), which was associated with impaired mitochondrial oxidative capacity as assessed by phosphorus magnetic resonance spectroscopy. Nishikawa et al. (11) showed that C57BL/6J mice that underwent partial Nx exhibited impaired mitochondrial biogenesis and reduced treadmill exercise capacity compared with sham-operated mice. Consistent with these clinical and preclinical studies, in the present study, we report that ADP-stimulated mitochondrial respiratory capacity was diminished (∼20%) in CKD mice regardless of the model, confirming a recent report in the adenine model (28). Taken together, these findings suggest that both adenine diet and 5/6 Nx models of CKD result in similar mitochondrial impairments in mice.
It has been established that the background genetic strain of mice can influence pathophysiology in numerous models of renal dysfunction (82–84). Indeed, C57BL6 mice, such as those used in this study, have been shown to be resistant to some aspects of CKD pathophysiology with the 5/6 Nx model, including cardiac fibrosis, proteinuria, glomerulosclerosis, and hypertension (82, 83, 85). Notably, the resistance displayed by the C57BL6 strain in the 5/6 Nx model does not appear in other models of renal dysfunction, including the adenine diet model (28), oxalate diet model (86), unilateral ureteral obstruction (84), and nephrotoxin treatment (87). Nonetheless, the results of the present study suggest that C57BL6 mice are not resistant to CKD-associated skeletal myopathy with the 5/6 Nx model, despite their relative resistance to some other CKD pathologies.
In conclusion, preclinical animal models of CKD are important tools required to understand the underlying mechanisms of renal disease as well as the pathophysiological consequences, including skeletal myopathy. Moreover, they serve as a critical research tool that allows testing of novel therapeutics that may translate to improve clinical outcomes in human patients with CKD. In the present study, we used two mouse models of CKD, adenine-induced nephropathy and 5/6 Nx, to compare their impact on the levels of uremic toxins, muscle mass, fiber CSA, and mitochondrial function. Both CKD models produced equivalent levels of renal dysfunction and uremia, which was accompanied near-identical reductions in skeletal muscle mass and mitochondrial respiratory function. Another significant finding in the present study was the striking sex differences in renal pathophysiology and muscle wasting between male and female mice. Notably, female mice displayed similar reductions in GFR compared with male mice but did not exhibit the same changes in BUN, serum Ca2+ or PTH levels, or muscle atrophy that male mice displayed. Considering that the prevalence of CKD is high in postmenopausal women (88) and that most preclinical CKD studies have been conducted in male animals (14, 15, 39), future studies should carefully consider the inclusion of female mice and possibilities of modeling the postmenopausal condition to better align clinically. Compared with the traditionally used 5/6 Nx model, the adenine model did not cause mortality and produced more consistent levels of GFR, BUN, and uremic toxin accumulation. Coupled with the ease of implementation, these findings support the use of the adenine diet model in mice for studies exploring adverse muscle impact.
SUPPLEMENTAL DATA
The data that support this study are available at https://doi.org/10.6084/m9.figshare.14317331.v2.
GRANTS
This work was supported by National Heart, Lung, and Blood Institute Grants R01HL149704 (to T.E.R.) and R01HL148597 (to S.T.S.), as well as a seed grant from the UF Office of Research (to T.E.R.). Salary support was also provided by American Heart Association Grant 18CDA34110044 (to T.E.R.).
DISCLAIMERS
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
K.K., S.T.S., and T.E.R. conceived and designed research; K.K., E.M.A., T.T., G.L., Z.R.S., and T.A.C. performed experiments; K.K., E.M.A., T.T., T.A.C., and T.E.R. analyzed data; K.K., E.M.A., T.T., G.L., K.A.O., S.T.S., and T.E.R. interpreted results of experiments; K.K. and T.E.R. prepared figures; K.K. and T.E.R. drafted manuscript; K.K., E.M.A., T.T., G.L., Z.R.S., T.A.C., K.A.O., S.T.S., and T.E.R. edited and revised manuscript; K.K., E.M.A., T.T., G.L., Z.R.S., T.A.C., K.A.O., S.T.S., and T.E.R. approved final version of manuscript.
ACKNOWLEDGMENTS
The authors thank Joy Cagmat, Dr. Timothy J. Garrett, and the Southeast Center for Integrated Metabolomics (http://secim.ufl.edu/) for assistance with metabolomics experiments.
REFERENCES
- 1.Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney disease. Lancet 389: 1238–1252, 2017. doi: 10.1016/S0140-6736(16)32064-5. [DOI] [PubMed] [Google Scholar]
- 2.Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis 41: 1–12, 2003. doi: 10.1053/ajkd.2003.50007. [DOI] [PubMed] [Google Scholar]
- 3.Gracia-Iguacel C, Gonzalez-Parra E, Perez-Gomez MV, Mahillo I, Egido J, Ortiz A, Carrero JJ. Prevalence of protein-energy wasting syndrome and its association with mortality in haemodialysis patients in a centre in Spain. Nefrologia 33: 495–505, 2013. doi: 10.3265/Nefrologia.pre2013.Apr.11979. [DOI] [PubMed] [Google Scholar]
- 4.Stenvinkel P, Heimburger O, Lindholm B. Wasting, but not malnutrition, predicts cardiovascular mortality in end-stage renal disease. Nephrol Dial Transplant 19: 2181–2183, 2004. doi: 10.1093/ndt/gfh296. [DOI] [PubMed] [Google Scholar]
- 5.Peng H, Cao J, Yu R, Danesh F, Wang Y, Mitch WE, Xu J, Hu Z. CKD stimulates muscle protein loss via Rho-associated protein kinase 1 activation. J Am Soc Nephrol 27: 509–519, 2016. doi: 10.1681/ASN.2014121208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Thomas SS, Mitch WE. Mechanisms stimulating muscle wasting in chronic kidney disease: the roles of the ubiquitin-proteasome system and myostatin. Clin Exp Nephrol 17: 174–182, 2013. doi: 10.1007/s10157-012-0729-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wang XH, Mitch WE. Mechanisms of muscle wasting in chronic kidney disease. Nat Rev Nephrol 10: 504–516, 2014. doi: 10.1038/nrneph.2014.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Avin KG, Chen NX, Organ JM, Zarse C, O'Neill K, Conway RG, Konrad RJ, Bacallao RL, Allen MR, Moe SM. Skeletal muscle regeneration and oxidative stress are altered in chronic kidney disease. PLoS One 11: e0159411, 2016. doi: 10.1371/journal.pone.0159411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gamboa JL, Billings FTt, Bojanowski MT, Gilliam LA, Yu C, Roshanravan B, Roberts LJ, 2nd, Himmelfarb J, Ikizler TA, Brown NJ. Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease. Physiol Rep 4: e12780, 2016. doi: 10.14814/phy2.12780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hallan S, Afkarian M, Zelnick LR, Kestenbaum B, Sharma S, Saito R, Darshi M, Barding G, Raftery D, Ju W, Kretzler M, Sharma K, de Boer IH. Metabolomics and gene expression analysis reveal down-regulation of the citric acid (TCA) cycle in non-diabetic CKD patients. EBioMedicine 26: 68–77, 2017. doi: 10.1016/j.ebiom.2017.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Nishikawa M, Ishimori N, Takada S, Saito A, Kadoguchi T, Furihata T, Fukushima A, Matsushima S, Yokota T, Kinugawa S, Tsutsui H. AST-120 ameliorates lowered exercise capacity and mitochondrial biogenesis in the skeletal muscle from mice with chronic kidney disease via reducing oxidative stress. Nephrol Dial Transplant 30: 934–942, 2015. doi: 10.1093/ndt/gfv103. [DOI] [PubMed] [Google Scholar]
- 12.Rao M, Jaber BL, Balakrishnan VS. Chronic kidney disease and acquired mitochondrial myopathy. Curr Opin Nephrol Hypertens 27: 113–120, 2018. doi: 10.1097/MNH.0000000000000393. [DOI] [PubMed] [Google Scholar]
- 13.Roshanravan B, Kestenbaum B, Gamboa J, Jubrias SA, Ayers E, Curtin L, Himmelfarb J, de Boer IH, Conley KE. CKD and muscle mitochondrial energetics. Am J Kidney Dis 68: 658–659, 2016. doi: 10.1053/j.ajkd.2016.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Berru FN, Gray SE, Thome T, Kumar RA, Salyers ZR, Coleman M, Dennis L, O'Malley K, Ferreira LF, Berceli SA, Scali ST, Ryan TE. Chronic kidney disease exacerbates ischemic limb myopathy in mice via altered mitochondrial energetics. Sci Rep 9: 15547, 2019. doi: 10.1038/s41598-019-52107-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sato E, Mori T, Mishima E, Suzuki A, Sugawara S, Kurasawa N, Saigusa D, Miura D, Morikawa-Ichinose T, Saito R, Oba-Yabana I, Oe Y, Kisu K, Naganuma E, Koizumi K, Mokudai T, Niwano Y, Kudo T, Suzuki C, Takahashi N, Sato H, Abe T, Niwa T, Ito S. Metabolic alterations by indoxyl sulfate in skeletal muscle induce uremic sarcopenia in chronic kidney disease. Sci Rep 6: 36618, 2016. doi: 10.1038/srep36618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Thome T, Salyers ZR, Kumar RA, Hahn D, Berru FN, Ferreira LF, Scali ST, Ryan TE. Uremic metabolites impair skeletal muscle mitochondrial energetics through disruption of the electron transport system and matrix dehydrogenase activity. Am J Physiol Cell Physiol 317: C701–C713, 2019. doi: 10.1152/ajpcell.00098.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mazumder MK, Giri A, Kumar S, Borah A. A highly reproducible mice model of chronic kidney disease: evidences of behavioural abnormalities and blood-brain barrier disruption. Life Sci 161: 27–36, 2016. doi: 10.1016/j.lfs.2016.07.020. [DOI] [PubMed] [Google Scholar]
- 18.Shimamura T, Morrison AB. A progressive glomerulosclerosis occurring in partial five-sixths nephrectomized rats. Am J Pathol 79: 95–106, 1975. [PMC free article] [PubMed] [Google Scholar]
- 19.Morrison AB. Experimentally induced chronic renal insufficiency in the rat. Lab Invest 11: 321–332, 1962. [PubMed] [Google Scholar]
- 20.Wang X, Chaudhry MA, Nie Y, Xie Z, Shapiro JI, Liu J. A mouse 5/6th nephrectomy model that induces experimental uremic cardiomyopathy. J Vis Exp 2017: 55825, 2017. doi: 10.3791/55825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Diwan V, Brown L, Gobe GC. Adenine-induced chronic kidney disease in rats. Nephrology (Carlton) 23: 5–11, 2018. doi: 10.1111/nep.13180. [DOI] [PubMed] [Google Scholar]
- 22.Diwan V, Small D, Kauter K, Gobe GC, Brown L. Gender differences in adenine-induced chronic kidney disease and cardiovascular complications in rats. Am J Physiol Renal Physiol 307: F1169–F1178, 2014. doi: 10.1152/ajprenal.00676.2013. [DOI] [PubMed] [Google Scholar]
- 23.Yokozawa T, Oura H, Okada T. Metabolic effects of dietary purine in rats. J Nutr Sci Vitaminol (Tokyo) 28: 519–526, 1982. doi: 10.3177/jnsv.28.519. [DOI] [PubMed] [Google Scholar]
- 24.Yokozawa T, Zheng PD, Oura H, Koizumi F. Animal model of adenine-induced chronic renal failure in rats. Nephron 44: 230–234, 1986. doi: 10.1159/000183992. [DOI] [PubMed] [Google Scholar]
- 25.Klinkhammer BM, Djudjaj S, Kunter U, Palsson R, Edvardsson VO, Wiech T, Thorsteinsdottir M, Hardarson S, Foresto-Neto O, Mulay SR, Moeller MJ, Jahnen-Dechent W, Floege J, Anders HJ, Boor P. Cellular and molecular mechanisms of kidney injury in 2,8-dihydroxyadenine nephropathy. J Am Soc Nephrol 31: 799–816, 2020. doi: 10.1681/ASN.2019080827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jia T, Olauson H, Lindberg K, Amin R, Edvardsson K, Lindholm B, Andersson G, Wernerson A, Sabbagh Y, Schiavi S, Larsson TE. A novel model of adenine-induced tubulointerstitial nephropathy in mice. BMC Nephrol 14: 116, 2013. doi: 10.1186/1471-2369-14-116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kieswich JE, Chen J, Alliouachene S, Caton PW, McCafferty K, Thiemermann C, Yaqoob MM. A novel model of reno-cardiac syndrome in the C57BL/6 mouse strain. BMC Nephrol 19: 346, 2018. doi: 10.1186/s12882-018-1155-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Thome T, Kumar RA, Burke SK, Khattri RB, Salyers ZR, Kelley RC, Coleman MD, Christou DD, Hepple RT, Scali ST, Ferreira LF, Ryan TE. Impaired muscle mitochondrial energetics is associated with uremic metabolite accumulation in chronic kidney disease. JCI Insight 6: e139826, 2021. doi: 10.1172/jci.insight.139826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ali BH, Adham SA, Al Za'abi M, Waly MI, Yasin J, Nemmar A, Schupp N. Ameliorative effect of chrysin on adenine-induced chronic kidney disease in rats. PLoS One 10: e0125285, 2015. doi: 10.1371/journal.pone.0125285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ali BH, Al Za'abi M, Adham SA, Al Suleimani Y, Karaca T, Manoj P, Al Kalbani J, Yasin J, Nemmar A. The effect of sildenafil on rats with adenine-induced chronic kidney disease. Biomed Pharmacother 108: 391–402, 2018. doi: 10.1016/j.biopha.2018.09.061. [DOI] [PubMed] [Google Scholar]
- 31.Donner AJ, Bell TA, Greenlee S, Graham MJ, Crooke RM. Characterization of the activity and Distribution of a 2'-O-Methoxyethyl-Modified Antisense Oligonucleotide in Models of Acute and Chronic Kidney Disease. Nucleic Acid Ther 28: 297–306, 2018. doi: 10.1089/nat.2018.0723. [DOI] [PubMed] [Google Scholar]
- 32.Schiffrin EL, Lipman ML, Mann JF. Chronic kidney disease: effects on the cardiovascular system. Circulation 116: 85–97, 2007. doi: 10.1161/CIRCULATIONAHA.106.678342. [DOI] [PubMed] [Google Scholar]
- 33.Qi Z, Whitt I, Mehta A, Jin J, Zhao M, Harris RC, Fogo AB, Breyer MD. Serial determination of glomerular filtration rate in conscious mice using FITC-inulin clearance. Am J Physiol Renal Physiol 286: F590–F596, 2004. doi: 10.1152/ajprenal.00324.2003. [DOI] [PubMed] [Google Scholar]
- 34.Rieg T. A High-throughput method for measurement of glomerular filtration rate in conscious mice. J Vis Exp 2013: e50330, 2013. doi: 10.3791/50330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Thome T, Kumar RA, Burke SK, Khattri RB, Salyers ZR, Kelley RC, Coleman MD, Christou DD, Hepple RT, Scali ST, Ferreira LF, Ryan TE. Impaired muscle mitochondrial energetics is associated with uremic metabolite accumulation in chronic kidney disease. JCI Insight 6: e139826, 2020. doi: 10.1172/jci.insight.139826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mayeuf-Louchart A, Hardy D, Thorel Q, Roux P, Gueniot L, Briand D, Mazeraud A, Bougle A, Shorte SL, Staels B, Chretien F, Duez H, Danckaert A. MuscleJ: a high-content analysis method to study skeletal muscle with a new Fiji tool. Skelet Muscle 8: 25, 2018. doi: 10.1186/s13395-018-0171-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ryan TE, Schmidt CA, Green TD, Spangenburg EE, Neufer PD, McClung JM. Targeted expression of catalase to mitochondria protects against ischemic myopathy in high fat fed mice. Diabetes 65: 2553–2568, 2016. doi: 10.2337/db16-0387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Duranton F, Cohen G, De Smet R, Rodriguez M, Jankowski J, Vanholder R, Argiles A; European Uremic Toxin Work Group. Normal and pathologic concentrations of uremic toxins. J Am Soc Nephrol 23: 1258–1270, 2012. [Erratum in J Am Soc Nephrol 24: 2127–2129, 2013]. doi: 10.1681/ASN.2011121175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Enoki Y, Watanabe H, Arake R, Sugimoto R, Imafuku T, Tominaga Y, Ishima Y, Kotani S, Nakajima M, Tanaka M, Matsushita K, Fukagawa M, Otagiri M, Maruyama T. Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1. Sci Rep 6: 32084, 2016. doi: 10.1038/srep32084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Harrison AP, Nielsen AH, Eidemak I, Molsted S, Bartels EM. The uremic environment and muscle dysfunction in man and rat. Nephron Physiol 103: p33–p42, 2006. doi: 10.1159/000090221. [DOI] [PubMed] [Google Scholar]
- 41.Koppe L, Pillon NJ, Vella RE, Croze ML, Pelletier CC, Chambert S, Massy Z, Glorieux G, Vanholder R, Dugenet Y, Soula HA, Fouque D, Soulage CO. p-Cresyl sulfate promotes insulin resistance associated with CKD. J Am Soc Nephrol 24: 88–99, 2013. doi: 10.1681/ASN.2012050503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vanholder R, De Smet R, Glorieux G, Argiles A, Baurmeister U, Brunet P, Clark W, Cohen G, De Deyn PP, Deppisch R, Descamps-Latscha B, Henle T, Jorres A, Lemke HD, Massy ZA, Passlick-Deetjen J, Rodriguez M, Stegmayr B, Stenvinkel P, Tetta C, Wanner C, Zidek W; European Uremic Toxin Work Group (EUTox) . Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 63: 1934–1943, 2003. doi: 10.1046/j.1523-1755.2003.00924.x. [DOI] [PubMed] [Google Scholar]
- 43.Vanholder R, Glorieux G, De Smet R, Lameire N; European Uremic Toxin Work Group (EUTox) . New insights in uremic toxins. Kidney Int Suppl 63: S6–S10, 2003. doi: 10.1046/j.1523-1755.63.s84.43.x. [DOI] [PubMed] [Google Scholar]
- 44.Bush KT, Singh P, Nigam SK. Gut-derived uremic toxin handling in vivo requires OAT-mediated tubular secretion in chronic kidney disease. JCI Insight 5: e133817, 2020. doi: 10.1172/jci.insight.133817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kikuchi H, Sasaki E, Nomura N, Mori T, Minamishima YA, Yoshizaki Y, Takahashi N, Furusho T, Arai Y, Mandai S, Yamashita T, Ando F, Maejima Y, Isobe K, Okado T, Rai T, Uchida S, Sohara E. Failure to sense energy depletion may be a novel therapeutic target in chronic kidney disease. Kidney Int 95: 123–137, 2019. doi: 10.1016/j.kint.2018.08.030. [DOI] [PubMed] [Google Scholar]
- 46.Dou L, Bertrand E, Cerini C, Faure V, Sampol J, Vanholder R, Berland Y, Brunet P. The uremic solutes p-cresol and indoxyl sulfate inhibit endothelial proliferation and wound repair. Kidney Int 65: 442–451, 2004. doi: 10.1111/j.1523-1755.2004.00399.x. [DOI] [PubMed] [Google Scholar]
- 47.Gava AL, Freitas FP, Balarini CM, Vasquez EC, Meyrelles SS. Effects of 5/6 nephrectomy on renal function and blood pressure in mice. Int J Physiol Pathophysiol Pharmacol 4: 167–173, 2012. [PMC free article] [PubMed] [Google Scholar]
- 48.Huang MC, Chen ME, Hung HC, Chen HC, Chang WT, Lee CH, Wu YY, Chiang HC, Hwang SJ. Inadequate energy and excess protein intakes may be associated with worsening renal function in chronic kidney disease. J Ren Nutr 18: 187–194, 2008. doi: 10.1053/j.jrn.2007.08.003. [DOI] [PubMed] [Google Scholar]
- 49.Cheung W, Yu PX, Little BM, Cone RD, Marks DL, Mak RH. Role of leptin and melanocortin signaling in uremia-associated cachexia. J Clin Invest 115: 1659–1665, 2005. doi: 10.1172/JCI22521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cheung WW, Kuo HJ, Markison S, Chen C, Foster AC, Marks DL, Mak RH. Peripheral administration of the melanocortin-4 receptor antagonist NBI-12i ameliorates uremia-associated cachexia in mice. J Am Soc Nephrol 18: 2517–2524, 2007. doi: 10.1681/ASN.2006091024. [DOI] [PubMed] [Google Scholar]
- 51.Kendrick J, Kestenbaum B, Chonchol M. Phosphate and cardiovascular disease. Adv Chronic Kidney Dis 18: 113–119, 2011. doi: 10.1053/j.ackd.2010.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Thomas L, Xue J, Murali SK, Fenton RA, Dominguez Rieg JA, Rieg T. Pharmacological Npt2a inhibition causes phosphaturia and reduces plasma phosphate in mice with normal and reduced kidney function. J Am Soc Nephrol 30: 2128–2139, 2019. doi: 10.1681/ASN.2018121250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Khattri RB, Thome T, Ryan TE. Tissue-specific 1H-NMR metabolomic profiling in mice with adenine-induced chronic kidney disease. Metabolites 11: 45, 2021. doi: 10.3390/metabo11010045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Tan RZ, Zhong X, Li JC, Zhang YW, Yan Y, Liao Y, Wen D, Diao H, Wang L, Shen HC. An optimized 5/6 nephrectomy mouse model based on unilateral kidney ligation and its application in renal fibrosis research. Ren Fail 41: 555–566, 2019. doi: 10.1080/0886022X.2019.1627220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kaminski TW, Pawlak K, Karbowska M, Mysliwiec M, Grzegorzewski W, Kuna J, Pawlak D. Association between uremic toxin-anthranilic acid and fibrinolytic system activity in predialysis patients at different stages of chronic kidney disease. Int Urol Nephrol 50: 127–135, 2018. doi: 10.1007/s11255-017-1729-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Moradi H, Sica DA, Kalantar-Zadeh K. Cardiovascular burden associated with uremic toxins in patients with chronic kidney disease. Am J Nephrol 38: 136–148, 2013. doi: 10.1159/000351758. [DOI] [PubMed] [Google Scholar]
- 57.Kaiser H, Parker E, Hamrick MW. Kynurenine signaling through the aryl hydrocarbon receptor: implications for aging and healthspan. Exp Gerontol 130: 110797, 2020. doi: 10.1016/j.exger.2019.110797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kaiser H, Yu K, Pandya C, Mendhe B, Isales CM, McGee-Lawrence ME, Johnson M, Fulzele S, Hamrick MW. Kynurenine, a tryptophan metabolite that increases with age, induces muscle atrophy and lipid peroxidation. Oxid Med Cell Longev 2019: 9894238, 2019. doi: 10.1155/2019/9894238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Popkov VA, Silachev DN, Zalevsky AO, Zorov DB, Plotnikov EY. Mitochondria as a source and a target for uremic toxins. Int J Mol Sci 20: 3094, 2019. doi: 10.3390/ijms20123094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Watanabe H, Miyamoto Y, Otagiri M, Maruyama T. Update on the pharmacokinetics and redox properties of protein-bound uremic toxins. J Pharm Sci 100: 3682–3695, 2011. doi: 10.1002/jps.22592. [DOI] [PubMed] [Google Scholar]
- 61.Deguchi T, Nakamura M, Tsutsumi Y, Suenaga A, Otagiri M. Pharmacokinetics and tissue distribution of uraemic indoxyl sulphate in rats. Biopharm Drug Dispos 24: 345–355, 2003. doi: 10.1002/bdd.370. [DOI] [PubMed] [Google Scholar]
- 62.Velenosi TJ, Hennop A, Feere DA, Tieu A, Kucey AS, Kyriacou P, McCuaig LE, Nevison SE, Kerr MA, Urquhart BL. Untargeted plasma and tissue metabolomics in rats with chronic kidney disease given AST-120. Sci Rep 6: 22526, 2016. doi: 10.1038/srep22526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bonanni A, Mannucci I, Verzola D, Sofia A, Saffioti S, Gianetta E, Garibotto G. Protein-energy wasting and mortality in chronic kidney disease. Int J Environ Res Public Health 8: 1631–1654, 2011. doi: 10.3390/ijerph8051631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Stenvinkel P, Carrero JJ, von Walden F, Ikizler TA, Nader GA. Muscle wasting in end-stage renal disease promulgates premature death: established, emerging and potential novel treatment strategies. Nephrol Dial Transplant 31: 1070–1077, 2016. doi: 10.1093/ndt/gfv122. [DOI] [PubMed] [Google Scholar]
- 65.Zhang L, Wang XH, Wang H, Du J, Mitch WE. Satellite cell dysfunction and impaired IGF-1 signaling cause CKD-induced muscle atrophy. J Am Soc Nephrol 21: 419–427, 2010. doi: 10.1681/ASN.2009060571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wang XH, Du J, Klein JD, Bailey JL, Mitch WE. Exercise ameliorates chronic kidney disease-induced defects in muscle protein metabolism and progenitor cell function. Kidney Int 76: 751–759, 2009. doi: 10.1038/ki.2009.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Du J, Wang X, Miereles C, Bailey JL, Debigare R, Zheng B, Price SR, Mitch WE. Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in catabolic conditions. J Clin Invest 113: 115–123, 2004. doi: 10.1172/JCI18330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhang LP, Pan J, Dong YJ, Tweardy DJ, Dong YL, Garibotto G, Mitch WE. Stat3 activation links a C/EBP delta to myostatin pathway to stimulate loss of muscle mass. Cell Metab 18: 368–379, 2013. doi: 10.1016/j.cmet.2013.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wang XNH, Hu ZY, Klein JD, Zhang LP, Fang FD, Mitch WE. Decreased miR-29 suppresses myogenesis in CKD. J Am Soc Nephrol 22: 2068–2076, 2011. doi: 10.1681/ASN.2010121278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vellanki K, Hou S. Menopause in CKD. Am J Kidney Dis 71: 710–719, 2018. doi: 10.1053/j.ajkd.2017.12.019. [DOI] [PubMed] [Google Scholar]
- 71.Collins BC, Laakkonen EK, Lowe DA. Aging of the musculoskeletal system: how the loss of estrogen impacts muscle strength. Bone 123: 137–144, 2019. doi: 10.1016/j.bone.2019.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Antus B, Hamar P, Kokeny G, Szollosi Z, Mucsi I, Nemes Z, Rosivall L. Estradiol is nephroprotective in the rat remnant kidney. Nephrol Dial Transplant 18: 54–61, 2003. doi: 10.1093/ndt/18.1.54. [DOI] [PubMed] [Google Scholar]
- 73.Guo Y, Sun N, Duan X, Xu X, Zheng L, Seriwatanachai D, Wang Y, Yuan Q. Estrogen deficiency leads to further bone loss in the mandible of CKD mice. PLoS One 11: e0148804, 2016. doi: 10.1371/journal.pone.0148804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Enns DL, Tiidus PM. The influence of estrogen on skeletal muscle: sex matters. Sports Med 40: 41–58, 2010. doi: 10.2165/11319760-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 75.Ikeda K, Horie-Inoue K, Inoue S. Functions of estrogen and estrogen receptor signaling on skeletal muscle. J Steroid Biochem 191: 105375, 2019. doi: 10.1016/j.jsbmb.2019.105375. [DOI] [PubMed] [Google Scholar]
- 76.Torres MJ, Kew KA, Ryan TE, Pennington ER, Lin CT, Buddo KA, Fix AM, Smith CA, Gilliam LA, Karvinen S, Lowe DA, Spangenburg EE, Zeczycki TN, Shaikh SR, Neufer PD. 17beta-estradiol directly lowers mitochondrial membrane microviscosity and improves bioenergetic function in skeletal muscle. Cell Metab 27: 167–179, 2018. doi: 10.1016/j.cmet.2017.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Torres MJ, Ryan TE, Lin CT, Zeczycki TN, Neufer PD. Impact of 17beta-estradiol on complex I kinetics and H2O2 production in liver and skeletal muscle mitochondria. J Biol Chem 293: 16889–16898, 2018. doi: 10.1074/jbc.RA118.005148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kitajima Y, Ono Y. Estrogens maintain skeletal muscle and satellite cell functions. J Endocrinol 229: 267–275, 2016. doi: 10.1530/JOE-15-0476. [DOI] [PubMed] [Google Scholar]
- 79.Gamboa JL, Roshanravan B, Towse T, Keller CA, Falck AM, Yu C, Frontera WR, Brown NJ, Ikizler TA. Skeletal muscle mitochondrial dysfunction is present in patients with CKD before initiation of maintenance hemodialysis. Clin J Am Soc Nephrol 15: 926–936, 2020. doi: 10.2215/CJN.10320819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kestenbaum B, Gamboa J, Liu S, Ali AS, Shankland E, Jue T, Giulivi C, Smith LR, Himmelfarb J, de Boer IH, Conley K, Roshanravan B. Impaired skeletal muscle mitochondrial bioenergetics and physical performance in chronic kidney disease. JCI Insight 5: e133289, 2020. doi: 10.1172/jci.insight.133289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sun CY, Cheng ML, Pan HC, Lee JH, Lee CC. Protein-bound uremic toxins impaired mitochondrial dynamics and functions. Oncotarget 8: 77722–77733, 2017. doi: 10.18632/oncotarget.20773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Leelahavanichkul A, Yan Q, Hu XZ, Eisner C, Huang YN, Chen R, Mizel D, Zhou H, Wright EC, Kopp JB, Schnermann J, Yuen PST, Star RA. Angiotensin II overcomes strain-dependent resistance of rapid CKD progression in a new remnant kidney mouse model. Kidney Int 78: 1136–1153, 2010. doi: 10.1038/ki.2010.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ma LJ, Fogo AB. Model of robust induction of glomerulosclerosis in mice: importance of genetic background. Kidney Int 64: 350–355, 2003. doi: 10.1046/j.1523-1755.2003.00058.x. [DOI] [PubMed] [Google Scholar]
- 84.Puri TS, Shakaib MI, Chang A, Mathew L, Olayinka O, Minto AWM, Sarav M, Hack BK, Quigg RJ. Chronic kidney disease induced in mice by reversible unilateral ureteral obstruction is dependent on genetic background. Am J Physiol Renal Physiol 298: F1024–F1032, 2010. doi: 10.1152/ajprenal.00384.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.O'Sullivan J, Finnie SL, Teenan O, Cairns C, Boyd A, Bailey MA, Thomson A, Hughes J, Benezech C, Conway BR, Denby L. Refining the mouse subtotal nephrectomy in male 129S2/SV mice for consistent modeling of progressive kidney disease with renal inflammation and cardiac dysfunction. Front Physiol 10: 1365, 2019. doi: 10.3389/fphys.2019.01365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mulay SR, Eberhard JN, Pfann V, Marschner JA, Darisipudi MN, Daniel C, Romoli S, Desai J, Grigorescu M, Kumar SV, Rathkolb B, Wolf E, de Angelis MH, Bauerle T, Dietel B, Wagner CA, Amann K, Eckardt KU, Aronson PS, Anders HJ, Knauf F. Oxalate-induced chronic kidney disease with its uremic and cardiovascular complications in C57BL/6 mice. Am J Physiol Renal Physiol 310: F785–F795, 2016. doi: 10.1152/ajprenal.00488.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ougaard MKE, Kvist PH, Jensen HE, Hess C, Rune I, Sondergaard H. Murine nephrotoxic nephritis as a model of chronic kidney disease. Int J Nephrol 2018: 8424502, 2018. doi: 10.1155/2018/8424502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bikbov B, Perico N, Remuzzi G; on behalf of the GBD Genitourinary Diseases Expert Group. Disparities in chronic kidney disease prevalence among males and females in 195 countries: analysis of the Global Burden of Disease 2016 Study. Nephron 139: 313–318, 2018. doi: 10.1159/000489897. [DOI] [PubMed] [Google Scholar]