Kidney Res Clin Pract > Epub ahead of print
Kim, Kim, Hyeon, Kim, Ye, Kim, Choi, Yi, Kim, Lee, and Lee: Obesity exacerbates ischemia-reperfusion injury and senescence in murine kidneys and perirenal adipose tissues

Abstract

Background

Obesity is a major worldwide health problem and can be related to cellular senescence. Along with the rise in obesity, the comorbidity of renal ischemia-reperfusion (IR) injury is increasing. Whether obesity accelerates the severity of IR injury and whether senescence contributes to these conditions remain unclear. We studied the degree of injury and cellular senescence in the IR kidneys and perirenal adipose tissues of high-fat-diet–induced obese mice.

Methods

C57BL/6 mice fed standard chow or a high-fat diet for 16 weeks were randomized to renal IR or sham group (n = 6–10 each). Renal IR was performed by unilateral clamping of the right renal pedicle for 30 minutes. Six weeks after surgery, renal function, perirenal fat/renal senescence, and histology were evaluated ex vivo.

Results

Obese mice showed more renal tubular damage and fibrosis in IR injury than control mice, even though the degree of ischemic insult was comparable. Renal expression of senescence and its secretory phenotype was upregulated in either IR injury or with a high-fat diet and was further increased in the IR kidneys of obese mice. Fat senescence and the expression of tumor necrosis factor alpha were also increased, especially in the perirenal depot of the IR kidneys, with a high-fat diet.

Conclusion

A high-fat diet aggravates IR injury in murine kidneys, which is associated, at least in part, with perirenal fat senescence and inflammation. These observations support the exploration of therapeutic targets of the adipo-renal axis in injured obese kidneys.

Introduction

Obesity, a major global health issue, is associated with an increased risk of the development of acute kidney injury as well as metabolic disorders such as diabetes and cardiovascular and kidney disease [1]. The comorbidity of acute kidney injury is increasing along with the rise in obesity. One-third of patients in intensive care units throughout the world have been reported to meet the criteria for obesity, and acute kidney injury may occur in over 50% of these patients [2,3]. Ischemia-reperfusion (IR) injury to the kidney is one of the most common causes of acute kidney injury [4]. Whether obesity accelerates the severity of IR injuries remains unclear.
Adipose tissue is a store of excess calories and is now recognized as an important endocrine organ [5]. This tissue synthesizes adipokines and secretes the adipokines via paracrine and endocrine pathways, thereby maintaining metabolic homeostasis. Moreover, visceral white adipose tissue, in particular, may play a role in the development of a variety of diseases [6,7]. Perirenal adipose tissue surrounds the kidneys, providing mechanical support. Recently, a close relationship has been demonstrated between perirenal adipose tissue and renal diseases, which is referred to as the adipo-renal axis [8,9].
Cellular senescence is an irreversible arrest of cell cycle progression evoked in response to stress and damage [10]. Obesity triggers renal senescence, suggesting a potential role in the progression of renal injury [11]. In obese mouse models, senescent cells accumulate in visceral adipose tissue [12]. However, the relationship between the kidney and senescence in perirenal adipose tissue remains unknown. In addition, whether renal IR injury affects adipose tissue around obese kidneys is uncertain.
Therefore, the aim of the study was to investigate whether obesity accelerates the severity of IR injury and cellular senescence in injured kidneys and perirenal adipose tissues in high-fat-diet–induced obese (DIO) mice.

Methods

All animal procedures followed the Guide for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Use Committee of the Pusan National University (No. 2021-004-A1C0(0)). C57BL male mice were fed for 16 weeks with either standard chow (control, CON) or a high-fat diet (Adjusted Calories Diet [60% Fat; TD.06414, 5.1 kcal/g; DooYeol Biotech] with approximately 15–25 kcal of daily calorie intake in each mouse) and randomized to either renal IR or sham groups. The CON_Sham, CON_IR, DIO_Sham, and DIO_IR groups comprised 6 to 10 mice each. Renal IR surgery was performed by unilateral clamping of the right renal pedicle for 30 minutes. Six weeks after surgery, urine samples were collected via cystocentesis, and mice were euthanized by terminal blood sampling (Fig. 1A). The kidneys were harvested, weighed, and halved for freezing and preservation in formalin. Perirenal fat (K) was collected from the lateral side of the kidneys, and epididymal fat (E) was obtained. Since mice fed with normal chow did not have enough perirenal fat to analyze, five fat groups other than perirenal fat were analyzed (Sham_E, DIO_Sham_E, DIO_Sham_K, DIO_IR_E, and DIO_IR_K).

Serum and urine measurements

The levels of serum and urine creatinine were assayed using enzyme-linked immunosorbent assays (ELISA) (#80350; Crystal Chem) according to the manufacturer’s instructions. Urinary microalbumin was measured by the Mouse Microalbumin ELISA Kit (#80630; Crystal Chem). Urine albumin excretion was then normalized to urine creatinine and expressed as urinary albumin-to-creatinine ratios (ACRs). The glucometer Accu-Chek Instant (Roche) was used for measuring glucose levels in whole blood after feeding [13].

Kidney and adipose tissue measurements

SYBR green-based quantitative reverse transcription (RT)-polymerase chain reaction (PCR) results were analyzed in frozen kidney and fat tissues, as described previously [14]. Briefly, total RNA was extracted and complementary DNA (cDNA) was generated. The PCR was performed with 100 ng of cDNA. The PCR was performed at 95 °C for 15 minutes, followed by 45 cycles at 95 °C for 20 seconds and 55 °C for 40 seconds. The PCR primer pairs, including those of Gapdh as an internal control, are shown in Supplementary Table 1 (available online). Fold changes of each target gene in the experimental groups relative to the control group were calculated using the 2-ΔΔCT method.
To estimate the degree of fibrosis, Masson-Trichrome staining was performed on renal paraffin sections. Interstitial fibrosis was semiautomatically quantified in 10 randomly chosen fields per section using ImageJ (FIJI software; National Institutes of Health) as previously described [15] and expressed as the percentage of blue-stained area to the total field area. Tubular injury in the outer medulla was scored in periodic acid‐Schiff (PAS) stained sections, as described [16]. To evaluate macrophage infiltration, renal paraffin sections were stained with F4/80 antibody (#ab111101; Abcam) [17]. For analysis of interstitial fibrosis and macrophage infiltration, the cortex area, including a few glomeruli, was randomly chosen, avoiding the perivascular regions as much as possible. To study the degree of senescence in renal and adipose tissue, renal and fat sections were stained for p21 antibody (#ab188224; Abcam). To assess proliferation and tumor necrosis factor alpha (TNF-α) expression in adipose tissue, paraffin-embedded sections were stained with Ki67 (#14-5698-82; Invitrogen) and TNF-α (#ab1793; Abcam) antibodies. F4/80+ macrophages, p21+ senescent cells, Ki67+ proliferative cells, and TNF-α+ cells were manually counted in 10 randomly chosen fields per section and expressed as positive cell numbers per field or a positive cell percentage out of the total cell number per field [18]. All slides were analyzed in a blinded manner.
Protein expression of p21 (1:1,000, #ab188224), TNF-α (1:1,000, #ab1793), and interleukin (IL)-6 (1:1,000, #ab208113; Abcam) was measured in 100 μg of protein from each homogenized adipose tissue sample by western blot. Beta-actin (1:5,000, #A3854; Sigma) was used as a loading control, and band intensity was quantified using ImageJ software.

In vitro study

HK-2 human renal tubular cells (Korean Cell Line Bank) were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco) containing 10% fetal bovine serum (Gibco), 60-U/mL penicillin G (Sigma), and 60-μg/mL streptomycin (Sigma). 3T3-L1 preadipocytes (Korean Cell Line Bank) were cultured in DMEM until a confluency of 70% was reached. To induce their differentiation into mature adipocytes, DMEM was changed to methylisobutylxanthine, dexamethasone, and insulin induction medium (0.5-mM IBMX and 1-μM dexamethasone; Abcam) that was replaced with insulin medium (10 μg/mL). All cultures were performed in a humidified 5% CO2 atmosphere at 37 °C. By days 7 to 10, fully differentiated adipocyte-like cells were obtained and tracked by Oil Red O staining using Oil Red O stock solution (MAK194; Sigma) to confirm lipid accumulation.
To induce senescence, two types of cells at a sub-confluent density were incubated with H2O2 (Fisher Scientific) at 0.2 mM for 48 hours; the untreated cells (in parallel) served as the control. H2O2 was removed 48 hours after treatment. Conditioned media were not replaced, but rather added for 7 days [19]. Cells were collected after 7 days, and conditioned media were harvested. The degree of senescence and senescence-associated secretory phenotype (SASP) in H2O2-treated cells was determined using RT-PCR, p21 staining (#ab188224), and ELISA. Briefly, SYBR green-based quantitative RT-PCR was analyzed in collected cells using the same methods as tissue PCR. The PCR primer pairs are shown in Supplementary Table 1 (available online). The levels of IL-1β (human ELISA kit, #88-7261-22; Invitrogen) and IL-6 (mouse ELISA kit, #M6000B; R&D system) in conditioned media were measured using ELISA according to the manufacturer’s instructions.
To determine a causal relationship, coculture experiments of H2O2-induced senescent adipocytes with normal renal tubular cells and H2O2-induced senescent renal tubular cells with normal adipocytes were performed on 6.5-mm Transwell plates with 3.0-µm Pore Polyester Membrane Inserts (#3472; Corning) [20]. At the start of the experiment, differentiated adipocytes and renal tubular cells were sown in the lower wells and treated with 0.2-mM H2O2 for 48 hours. The H2O2 was removed before the coculture. Untreated, opposite cells were sown in the upper wells. The upper wells were attached to the lower wells, and cells were cultured in two layers with medium communication between the wells for 7 days. In cocultured cells, RT-PCR for senescence and SASP genes was performed. The PCR primer pairs are shown in Supplementary Table 1 (available online).

Statistical analysis

Statistical analysis was performed using SPSS version 13.0 (SPSS Inc.). Parametric (independent t test) and nonparametric (Mann-Whitney U test) tests were used for comparisons between groups. Data from more than two groups were assessed by the analysis of variance and Kruskal-Wallis test. A p-value of ≤0.05 was considered statistically significant.

Results

The final body weights of DIO mice were approximately twice those of the control groups (all p ≤ 0.02) (Fig. 1B). The ratios of right to left kidney were decreased in IR compared to sham groups (CON_Sham vs. CON_IR, p = 0.001; CON_Sham vs. DIO_IR, p = 0.006; and DIO_Sham vs. DIO_IR, p = 0.04); but the ratio was similar between CON-IR and DIO-IR (p = 0.35) (Fig. 1C). Serum creatinine and blood glucose levels did not differ statistically among the groups (p = 0.23 and p = 0.12) (Fig. 1D, F). The urine ACR was increased in mice receiving a high-fat diet compared to control mice (all p ≤ 0.02) (Fig. 1E).
Renal gene expression of Cdkn1a, which translates p21, was upregulated in the IR or DIO groups (all p ≤ 0.002) and was further increased in DIO_IR kidneys (vs. CON_IR, p = 0.01) (Fig. 2A). Expression of Cdkn2a, p16, was upregulated only in IR injury groups compared to shams (all p ≤ 0.003) (Fig. 2A, (B). Secretory phenotype renal gene expression was upregulated in CON_IR or DIO groups (all p ≤ 0.02). Moreover, expression of Il1α, Mmp3, Il6, Tnfα, and Serpine1 was further increased in the IR kidneys of mice on a high-fat diet (vs. CON_IR, all p ≤ 0.03) (Fig. 2CE). The number of p21+ senescent cells was increased in either the IR or DIO groups (all p ≤ 0.006), and this number tended to be greater in DIO-IR than in control-IR (p = 0.07) (Fig. 2E, F). The degree of renal tubular injury was increased in IR groups compared to sham groups (all p ≤ 0.003) and was higher in DIO_IR than in CON_IR (p = 0.05) (Fig. 3A, D). PAS staining showed glomerulomegaly and vacuoles localized primarily to proximal tubular cells in DIO groups. Renal fibrosis was increased in either the IR or DIO groups (all p ≤ 0.004), and fibrosis tended to be greater in DIO-IR than in control-IR (p = 0.08) (Fig. 3B, E; Supplementary Fig. 1, available online). The difference in F4/80-positive macrophage infiltration was remarkable in IR compared to Sham groups (all p ≤ 0.05) but was not statistically different between the two IR groups, possibly due to high variability (p = 0.15) (Fig. 3C, F; Supplementary Fig. 2, available online).
Expression of the p21 gene was higher in the perirenal depots of mice on a high-fat diet with or without IR injury than in the control mice (p = 0.01 and p = 0.04, respectively) (Fig. 4A). p21 expression in fat sections was increased in mice on a high-fat diet compared to control mice (all p ≤ 0.04) and further upregulated in perirenal fat in DIO_IR mice than in the fat from other sources (all p ≤ 0.002) (Fig. 4B, C). The number of Ki67+ proliferative cells was increased in the epididymal and perirenal fat of mice on a high-fat diet regardless of IR injury compared to mice on a normal chow (all p ≤ 0.03). However, perirenal adipose tissue in obese mice with IR injury showed decreased Ki67+ cells compared to other adipose tissue depots in obese mice, implying upregulated senescence in this depot (all p ≤ 0.003) (Supplementary Fig. 1, available online). Adiponectin expression was downregulated in both epididymal and perirenal depots of mice on a high-fat diet compared to the controls (all p = 0.003), but was slightly higher in the perirenal depots of DIO_Sham and DIO_IR than in the epididymal depot of DIO_Sham (p = 0.05 and p = 0.04, respectively) (Fig. 5A). Tnfα and Il6 gene expression was upregulated only in the perirenal depots of mice on a high-fat diet compared to the epididymal depots of mice either on or not on a high-fat diet (all p ≤ 0.02). Tnfα gene expression in the epididymal fat tended to be higher in DIO_Sham than in Sham (p = 0.08), and TNF-α protein expression was higher in DIO_IR_K than in Sham_E and DIO_Sham_K (p = 0.02 and p = 0.04, respectively). IL-6 protein expression had a strong tendency to increase in DIO_IR_K compared to Sham_E (p = 0.06) (Fig. 5BE; Supplementary Fig. 2, available online). TNF-α positive cell percentage in fat sections was increased in mice on a high-fat diet compared to control mice (all p ≤ 0.01) and was further upregulated in perirenal fat in DIO_IR mice than in the fat from other sources (all p ≤ 0.04) (Fig. 5F, G).
In an in vitro study, HK2 cells and fully differentiated 3T3-L1 cells (Fig. 6A) represented renal tubular cells and mature adipocytes, respectively. Initially, senescence was induced by H2O2 in both cell lines. Then, cells were cultured in two layers with medium communication between wells for 7 days using Transwell plates (Fig. 6B). Most of the adipocytes and renal tubular cells incubated with H2O2 at 0.2 mM for 48 hours expressed p21 and were senescent (Fig. 6C, D). Conditioned medium harvested at day 7 after removal of H2O2 contained increased levels of IL-6 in the H2O2-treated adipocytes and IL-1β in the H2O2-treated tubular cells compared to control cells (p = 0.003 and p = 0.05, respectively) (Fig. 6E, F). Relative gene expression of Cdkn1a, Cdkn2a, Il1β, and Tnfα was higher in cocultured adipocytes with senescent renal tubular cells than with control ones (all p ≤ 0.05) (Fig. 6G). Cocultured renal tubular cells with senescent adipocytes also showed increased CDKN1A, CDKN2A, and IL1Β expression (all p ≤ 0.04) (Fig. 6H).

Discussion

In this study, obese mice showed more renal tubular damage and fibrosis with IR injury than control mice, even though the degree of ischemic insult was comparable. Renal expression of senescence and its secretory phenotype were upregulated in either IR injury or with a high-fat diet and further increased in the IR kidneys of obese mice. Fat senescence and TNF-α expression were also increased, especially in the perirenal depot of the IR kidneys of mice on a high-fat diet. Therefore, a high-fat diet exacerbates IR injury in murine kidneys, which may be associated with perirenal fat senescence and inflammation. These results support interactions between the injured kidney and perirenal adipose tissue and the potential role of senescence in this adipo-renal axis.
Previous studies have shown albuminuria without nephritis and the development of glomerulomegaly and glomerulosclerosis in obese patients, now recognized as obesity-related glomerulopathy [1]. Our mice on a high-fat diet encountered glomerulomegaly and glomerular damage demonstrated by albuminuria and upregulated senescence in the kidneys. Obesity triggers the accumulation of senescent cells in various organs and contributes to the development of obesity-related dysfunction [21,22]. In obesity, organ lipid deposition is associated with impaired fatty acid oxidation and mitochondrial dysfunction, leading to cellular senescence [19]. Mitochondrial dysfunction during cellular senescence is a key factor in the consolidation of senescence and the initiation of the secretory phenotype [22,23]. A recent study showed that renal cellular senescence is induced by obesity and dyslipidemia and correlates with renal dysfunction and hypoxia [11]. Furthermore, the clearance of senescent cells in obesity recapitulated functional and metabolic improvements [11,22].
Senescent cells exhibit a secretion phenotype in 30% to 70% of cases, which fosters their senescence and the senescence of adjacent cells and exerts deleterious effects locally and systemically [24]. Recent studies have demonstrated that transplantation of a small number of senescent cells can even provoke significant injury and dysfunction in various organs, including the kidney and the knee joint [25,26]. Our study showed that both obesity and IR injury elicited senescence in the kidney and perirenal adipose tissue. Moreover, the perirenal fat around the injured kidney was more senescent and proinflammatory, suggesting the potential role of the SASP in adipo-renal interactions. Although confirming a causal connection in our mouse study is difficult, we identified the propagation of senescence by SASP in vitro coculture experiments between renal tubular cells and adipocytes. Further studies are needed to confirm the direct cause-and-effect relationship in this exacerbation.
Perirenal adipose tissue consists of white adipocytes, inactive and active brown adipocytes, preadipocytes, sympathetic nerve endings, vascular structures, and various types of inflammatory cells [9]. This study demonstrated boosted senescence and TNF-α expression in the perirenal depot of the IR kidneys of mice on a high-fat diet. Recent studies showed that perirenal adipose tissue contributes to chronic renal pathology. Perirenal fat thickness is an independent risk factor for deterioration of the glomerular filtration rate in hypertensive patients and type 2 diabetes patients [27,28]. Increased perirenal fat mechanically compresses the kidney; raises intrarenal pressure; reduces renal blood flow; and, in turn, promotes renin secretion and tubular sodium reabsorption, which results in the acceleration of kidney disease progression [29]. Accumulation of perirenal adipose tissue releases adipokines including leptin and resistin, leading to the expression of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6 [9]. However, this accumulation reduces anti-inflammatory adiponectin release. These adipokines and cytokines may affect adjacent kidneys in a paracrine manner. Finally, perirenal adipose tissue is regarded as a reservoir of mesenchymal stem cells (MSCs), which retain regenerative and immunomodulatory capacity [30]. A recent study showed that obesity induces cellular senescence in adipose-tissue–derived MSCs and leads to impaired reparative properties [31]. Indeed, the IR kidneys were injured worse in our mice on a high-fat diet, implying damage to the endogenous MSC repair system after IR insult in obese mice.
We acknowledge some limitations in our study. The murine study is limited by small group sizes and the variability of the IR surgery. In obesity-related animal models, the effect of obesity may overlap with the impact of overt diabetes. However, our DIO mice did not show significantly increased blood glucose levels, so we could exclude the effect of diabetes on the kidney. We could not compare the perirenal fat of obese mice directly to mice with normal chow because of insufficient amounts of perirenal fat in normal-weighted mice. We included only perirenal fat collected from the lateral side of the kidneys, which is regarded as white adipose tissue. Thus, the effect of brown adipose tissue, which may be around the adrenal gland and renal sinus, on the kidney cannot be excluded. In our study, epididymal fat depots did not exhibit upregulation of proinflammatory cytokines including TNF-α and IL-6 in RT-PCR or western blotting, but TNF-α expression in immunohistochemistry staining was increased in the epididymal fat depot. This discrepancy between gene and protein expression might be secondary to differences in protein translation and degradation rates [32]. Also, due to the limited amount of murine perirenal adipose tissue, there may still be problems with handling in protein analysis. Epididymal fat also showed adipocyte hypertrophy, increased senescence, and downregulated adiponectin. Since different regional adipose tissues may express various inflammatory properties [33], further studies are needed to confirm the differences between the various regional adipose tissues. Finally, clamping the renal vessels in the IR surgery may damage perirenal adipocytes. Nevertheless, the perirenal adipose tissue is well-vascularized and supplied by an anastomosing capillary network, which is produced by the branches of the left colic, lower adrenal, renal, lumbar, and ovarian or testicular arteries [9].
In conclusion, IR injury in murine kidneys is exacerbated by a high-fat diet and is related, at least in part, to senescence and inflammation in perirenal fat. These results suggest the importance of interactions between the injured kidney and perirenal adipose tissue and the potential role of senescence in this adipo-renal axis. Further research is needed on therapeutic targets of the adipo-renal axis in injured obese kidneys.

Supplementary Materials

Supplementary data are available at Kidney Research and Clinical Practice online (https://doi.org/10.23876/j.krcp.22.193).

Notes

Conflicts of interest

All authors have no conflicts of interest to declare.

Funding

This study was supported by the Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, the Young Investigator Research Grant from the Korean Society of Nephrology (K-NRF-Chongkundang, 2020), and the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2020R1F1A1073317).

Data sharing statement

The data presented in this study are available upon reasonable request from the corresponding author.

Authors’ contributions

Conceptualization: SRK, IYK, SBL, DWL

Funding acquisition: SRK

Investigation, Methodology: SRK, YSK, MJK

Data curation, Formal analysis: YSK, JMH, SJK, BMY, MJK, SRK, BHC, DY

Supervision: IYK, SBL, DWL

Writing–original draft: SRK

Writing–review & editing: SRK, BHC, DY, IYK, SBL, DWL

All authors read and approved the final version of the manuscript.

Figure 1.

Characteristics of diet-induced obesity (DIO) mice with ischemia-reperfusion (IR) injury and serum and urine measurements.

(A) Experimental design. Mice fed for 16 weeks with standard chow (control, CON) or a high-fat diet were studied at 6 weeks after renal IR or sham surgery. (B) Final body weights. (C) Right kidney (RK) and left kidney (LK) weights and their ratios in each group. (D) Serum creatinine levels. (E) Blood glucose levels. (F) Urine albumin-to-creatinine ratios (ACR). *p ≤ 0.05 vs. CON_Sham, p ≤ 0.05 vs. CON_IR, p ≤ 0.05 vs. DIO_Sham.
NS, no significance.
j-krcp-22-193f1.jpg
Figure 2.

Senescence in the ischemia-reperfusion (IR) injured kidneys of diet-induced obese (DIO) mice.

(A–D) Renal gene expression of senescence and its secretory phenotype by real-time polymerase chain reaction (relative to Gapdh). (E) The number of p21-positive senescent cells counted manually per the field. (F) Representative renal images of p21 immunohistochemistry staining. *p ≤ 0.05 vs. CON_Sham, p ≤ 0.05 vs. CON_IR, p ≤ 0.05 vs. DIO_Sham.
CON, control.
j-krcp-22-193f2.jpg
Figure 3.

Renal tubular injury, fibrosis, and inflammation in the ischemia-reperfusion (IR) injured kidneys of diet-induced obese (DIO) mice.

(A–C) Representative renal images of periodic acid-Schiff (PAS), Masson’s trichrome (MT), and F4/80 staining. (D) Renal tubular injury scores as the quantification of PAS staining. (E) Renal fibrosis area percentage as the quantification of MT staining. (F) The number of F4/80-positive macrophages counted manually per the field. *p ≤ 0.05 vs. CON_Sham, p ≤ 0.05 vs. CON_IR, p ≤ 0.05 vs. DIO_Sham.
CON, control.
j-krcp-22-193f3.jpg
Figure 4.

Senescence expression in the epididymal (E) and perirenal (K) fat depots of mice undergoing sham or ischemia-reperfusion (IR) injury with normal chow or a high-fat diet (diet-induced obesity, DIO).

(A) Cdkn1a gene expression by real-time polymerase chain reaction (relative to Gapdh). (B, C) p21 immunohistochemistry (IHC) staining and its quantification. Arrows, p21-positive cell. *p ≤ 0.05 vs. Sham_E, p ≤ 0.05 vs. DIO_Sham_E, p ≤ 0.05 vs. DIO_Sham_K; §p ≤ 0.05 vs. DIO_IR_E.
j-krcp-22-193f4.jpg
Figure 5.

Expression of adiponectin and cytokines in the epididymal (E) and perirenal (K) fat depots of mice undergoing sham or ischemia-reperfusion (IR) injury with normal chow or a high-fat diet (diet-induced obesity, DIO).

(A–C) Adiponectin, TNFα, and Il6 gene expression by real-time polymerase chain reaction (relative to Gapdh). (D, E) TNF-α and IL-6 protein expression by western blotting (relative to β-actin). (F, G) The quantification of TNF-α–positive cell percentage (out of total cell number) in the field and representative images of TNF-α immunohistochemistry (IHC) staining. Arrows, TNF-α–positive cell. *p ≤ 0.05 vs. Sham_E, p ≤ 0.05 vs. DIO_Sham_E, p ≤ 0.05 vs. DIO_Sham_K, §p ≤ 0.05 vs. DIO_IR_E.
IL, interleukin; NS, no significance; TNF-α, tumor necrosis factor alpha.
j-krcp-22-193f5.jpg
Figure 6.

In vitro study of the coculture system using adipocytes (3T3-L1s) and renal tubular cells (HK2s).

(A) Representative image of Oil Red O staining in fully differentiated adipocyte-like cells from 3T3-L1s. (B) Experimental design. Senescence was induced by H2O2 in adipocytes and renal tubular cells, respectively. Using Transwell plates, cells were cultured in two layers, with H2O2-induced senescent adipocytes in the lower wells and control tubular cells in the upper wells, or with H2O2-induced senescent tubular cells in the lower wells and control adipocytes in the upper wells. (C, D) p21 immunohistochemistry staining images (×200) in H2O2-treated adipocytes and tubular cells, showing H2O2-induced senescence. (E) interleukin (IL)-6 level in conditioned medium from H2O2-treated adipocytes, measured by enzyme-linked immunosorbent assay. (F) IL-1β level in conditioned medium from H2O2-treated tubular cells. (G) Senescence and its secretory phenotype gene expression by real-time polymerase chain reaction (relative to Gapdh) in cocultured adipocytes with H2O2-treated tubular cells. (H) Senescence and its secretory phenotype gene expression in cocultured tubular cells with H2O2-treated adipocytes. *p ≤ 0.05 vs. control.
DAPI, 4',6-diamidino-2-phenylindole.
j-krcp-22-193f6.jpg

References

1. Kanasaki K, Kitada M, Kanasaki M, Koya D. The biological consequence of obesity on the kidney. Nephrol Dial Transplant 2013;28 Suppl 4:iv1–iv7.
crossref pmid
2. Schiffl H, Lang SM. Obesity, acute kidney injury and outcome of critical illness. Int Urol Nephrol 2017;49:461–466.
crossref pmid pdf
3. Schiffl H. Obesity and the survival of critically ill patients with acute kidney injury: a paradox within the paradox? Kidney Dis (Basel) 2020;6:13–21.
crossref pmid pdf
4. Bonventre JV, Weinberg JM. Recent advances in the pathophysiology of ischemic acute renal failure. J Am Soc Nephrol 2003;14:2199–2210.
crossref pmid
5. Hammoud SH, AlZaim I, Al-Dhaheri Y, Eid AH, El-Yazbi AF. Perirenal adipose tissue inflammation: novel insights linking metabolic dysfunction to renal diseases. Front Endocrinol (Lausanne) 2021;12:707126.
crossref pmid pmc
6. Stieber C, Malka K, Boucher JM, Liaw L. Human perivascular adipose tissue as a regulator of the vascular microenvironment and diseases of the coronary artery and aorta. J Cardiol Cardiovasc Sci 2019;3:10–15.
crossref pmid pmc
7. Schaffer JE. Lipotoxicity: when tissues overeat. Curr Opin Lipidol 2003;14:281–287.
crossref pmid
8. Hua Y, Herder C, Kalhoff H, et al. Inflammatory mediators in the adipo-renal axis: leptin, adiponectin, and soluble ICAM-1. Am J Physiol Renal Physiol 2020;319:F469–F475.
crossref pmid
9. Grigoraș A, Balan RA, Căruntu ID, et al. Perirenal adipose tissue-current knowledge and future opportunities. J Clin Med 2021;10:1291.
crossref pmid pmc
10. Kim SR, Puranik AS, Jiang K, et al. Progressive cellular senescence mediates renal dysfunction in ischemic nephropathy. J Am Soc Nephrol 2021;32:1987–2004.
crossref pmid pmc
11. Kim SR, Jiang K, Ogrodnik M, et al. Increased renal cellular senescence in murine high-fat diet: effect of the senolytic drug quercetin. Transl Res 2019;213:112–123.
crossref pmid pmc
12. Palmer AK, Xu M, Zhu Y, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019;18:e12950.
pmid pmc
13. Morley LA, Gomez TH, Goldman JL, Flores R, Robinson MA. Accuracy of 5 point-of-care glucometers in C57BL/6J mice. J Am Assoc Lab Anim Sci 2018;57:44–50.
pmid pmc
14. Xiang DM, Song XZ, Zhou ZM, et al. Chronic kidney disease promotes chronic inflammation in visceral white adipose tissue. Am J Physiol Renal Physiol 2017;312:F689–F701.
crossref pmid
15. Crowe AR, Yue W. Semi-quantitative determination of protein expression using immunohistochemistry staining and analysis: an integrated protocol. Bio Protoc 2019;9:e3465.
crossref pmid pmc
16. Kim SR, Jiang K, Chen X, et al. Selective kidney targeting increases the efficacy of mesenchymal stromal/stem cells for alleviation of murine stenotic-kidney senescence and damage. J Tissue Eng Regen Med 2022;16:550–558.
crossref pmid pmc pdf
17. Kim SR, Zou X, Tang H, et al. Increased cellular senescence in the murine and human stenotic kidney: effect of mesenchymal stem cells. J Cell Physiol 2021;236:1332–1344.
crossref pmid pdf
18. Kim SR, Eirin A, Zhang X, Lerman A, Lerman LO. Mitochondrial protection partly mitigates kidney cellular senescence in swine atherosclerotic renal artery stenosis. Cell Physiol Biochem 2019;52:617–632.
crossref pmid pdf
19. Rattananinsruang P, Noonin C, Yoodee S, Thongboonkerd V. Comparative analysis of markers for H2O2-induced senescence in renal tubular cells. Environ Toxicol Pharmacol 2023;97:104039.
crossref pmid
20. Ichikawa J, Okada A, Taguchi K, et al. Increased crystal-cell interaction in vitro under co-culture of renal tubular cells and adipocytes by in vitro co-culture paracrine systems simulating metabolic syndrome. Urolithiasis 2014;42:17–28.
crossref pmid pdf
21. Ogrodnik M, Miwa S, Tchkonia T, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 2017;8:15691.
crossref pmid pmc pdf
22. Ogrodnik M, Zhu Y, Langhi LG, et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab 2019;29:1061–1077.
crossref pmid pmc
23. Passos JF, Nelson G, Wang C, et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol Syst Biol 2010;6:347.
crossref pmid pmc pdf
24. Xu M, Pirtskhalava T, Farr JN, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 2018;24:1246–1256.
pmid pmc
25. Kim SR, Jiang K, Ferguson CM, et al. Transplanted senescent renal scattered tubular-like cells induce injury in the mouse kidney. Am J Physiol Renal Physiol 2020;318:F1167–F1176.
crossref pmid pmc
26. Xu M, Bradley EW, Weivoda MM, et al. Transplanted senescent cells induce an osteoarthritis-like condition in mice. J Gerontol A Biol Sci Med Sci 2017;72:780–785.
crossref pmid
27. Lamacchia O, Nicastro V, Camarchio D, et al. Para- and perirenal fat thickness is an independent predictor of chronic kidney disease, increased renal resistance index and hyperuricaemia in type-2 diabetic patients. Nephrol Dial Transplant 2011;26:892–898.
crossref pmid
28. Geraci G, Zammuto MM, Mattina A, et al. Para-perirenal distribution of body fat is associated with reduced glomerular filtration rate regardless of other indices of adiposity in hypertensive patients. J Clin Hypertens (Greenwich) 2018;20:1438–1446.
crossref pmid pmc pdf
29. Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 2019;15:367–385.
crossref pmid pmc pdf
30. Baer PC, Koch B, Hickmann E, et al. Isolation, characterization, differentiation and immunomodulatory capacity of mesenchymal stromal/stem cells from human perirenal adipose tissue. Cells 2019;8:1346.
crossref
31. Klomjit N, Conley SM, Zhu XY, et al. Effects of obesity on reparative function of human adipose tissue-derived mesenchymal stem cells on ischemic murine kidneys. Int J Obes (Lond) 2022;46:1222–1233.
crossref pmid pmc pdf
32. Raj A, Peskin CS, Tranchina D, Vargas DY, Tyagi S. Stochastic mRNA synthesis in mammalian cells. PLoS Biol 2006;4:e309.
crossref pmid pmc
33. Gruzdeva O, Borodkina D, Uchasova E, Dyleva Y, Barbarash O. Localization of fat depots and cardiovascular risk. Lipids Health Dis 2018;17:218.
crossref pmid pmc pdf


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2024 by The Korean Society of Nephrology.

Developed in M2PI

Close layer