Skip to main content

Effects of xenogeneic transplantation of umbilical cord-derived mesenchymal stem cells combined with irbesartan on renal podocyte damage in diabetic rats

Abstract

Background

The leading cause of end-stage renal disease (ESRD) is diabetic nephropathy (DN). Podocyte damage is an early event in the development of DN. Currently, there is no effective treatment strategy that can slow the progression of DN or reverse its onset. The role of mesenchymal stem cells (MSCs) transplantation in diabetes and its complications has been extensively studied, and diabetic nephropathy has been a major focus. Irbesartan exerts reno-protective effects independent of lowering blood pressure, can reduce the incidence of proteinuria in rats, and is widely used clinically. However, it remains undetermined whether the combined utilization of the angiotensin II receptor antagonist irbesartan and MSCs could enhance efficacy in addressing DN.

Methods

A commonly used method for modeling type 2 diabetic nephropathy (T2DN) was established using a high-fat diet and a single low-dose injection of STZ (35 mg/kg). The animals were divided into the following 5 groups: (1) the control group (CON), (2) the diabetic nephropathy group (DN), (3) the mesenchymal stem cells treatment group (MSCs), (4) the irbesartan treatment group (Irb), and (5) the combined administration group (MSC + Irb). MSCs (2 × 106 cells/rat) were injected every 10 days through the tail vein for a total of three injections; irbesartan (30 mg/kg/d) was administered by gavage. Additionally, the safety and homing of mesenchymal stem cells were verified using positron emission tomography (PET) imaging.

Results

The combination treatment significantly reduced the UACR, kidney index, IGPTT, HOMA-IR, BUN, serum creatine, and related inflammatory factor levels and significantly improved renal function parameters and the expression of proteins related to glomerular podocyte injury in rats. Moreover, MSCs can homing target to damaged kidneys.

Conclusions

Compared to the administration of MSCs or irbesartan alone, the combination of MSCs and irbesartan exerted better protective effects on glomerular podocyte injury, providing new ideas for the clinical application of mesenchymal stem cells.

Introduction

Diabetes mellitus (DM) is an intractable global health challenge, and the number of people suffering from this disease reached 451 million worldwide as of 2017 and is projected to increase to 552 million by 2030 [1]. Approximately 20–40% of people with diabetes suffer from diabetic nephropathy, particularly those with type 2 diabetes mellitus (T2DM) [2].

Diabetic nephropathy (DN) is a type of kidney disease caused by chronic high blood glucose and is one of the most common and serious complications of diabetes. In the early stages of diabetic nephropathy, the glomeruli are damaged, and microalbumin is present in the urine, which is often one of the first signs. If not controlled in time, the patient's condition will rapidly deteriorate, and the patient will eventually develop end-stage renal disease (ESRD) and require hemodialysis or even kidney transplantation [3].

The pathogenesis of DN has not been fully elucidated, but the role of chronic inflammation induced by prolonged hyperglycemia in podocyte injury has received increasing attention. Podocytes are major components of the glomerular filtration barrier (GFB) [4], thus, podocyte injury impairs GFB integrity and is strongly associated with diffuse glomerulosclerosis and ESRD [5].

A hyperglycemic environment can increase the release of many cytokines, such as tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6). Increased levels of these cytokines can promote the development of kidney inflammation, which can disrupt podocyte structure and reduce the number of podocytes. In turn, changes in podocyte structure and a reduction in podocyte numbers can lead to proteinuria [6]. To better understand the relationship between inflammation and DN, we used Rat Cytokine arrays (Catalog #: GSR-CYT-3-1, RayBiotech Inc) to confirm the expression of 27 cytokines in the renal cortex of rats. The advantages of antibiotic microarrays include low sample consumption, high sensitivity, and fast output of experimental results.

Recently, stem cell therapy, as a potential regenerative treatment [7], has been widely applied in the research of diabetes and its complications, especially DN [8]. Mesenchymal stem cells (MSCs) were initially isolated and characterized from bone marrow but were subsequently isolated from other tissues, such as adipose tissue, placenta, and umbilical cord blood. Among these, umbilical cord-derived mesenchymal stem cells (UC-MSCs) are used as an alternative source of stem cells to bone marrow due to their advantages such as easy accessibility, low immunogenicity, and lack of ethical concerns [9]. Besides, MSCs can also migrate to damaged sites, regenerate tissue [10, 11] and secrete paracrine mediators [12]. They also have excellent anti-inflammatory [13] and immunosuppressive [14] properties, making them safe for allogeneic and xenogeneic infusion. Recent studies [15, 16] have proved that cell-based therapy endured a dose-dependent effect (a single dose was inferior to two doses) on improving the outcomes in damaged organs.

Since angiotensin II is the primary regulator of glomerular filtration, current guidelines recommend the use of renin-angiotensin-aldosterone system (RAAS) inhibitors as the primary treatment for DN to reduce glomerular pressure and increase the filtration rate [17]. Among these agents, the angiotensin II receptor antagonist irbesartan can effectively prevent the progression of type 2 diabetic nephropathy (T2DN), and this protective effect is independent of its ability to reduce blood pressure. In a clinical trial [18], compared with amlodipine, a placebo, and other antihypertensive drugs, irbesartan was associated with better renal outcomes and lower rates of other adverse effects, including cardiovascular mortality, than the placebo.

Previous studies showed that combined treatment with mesenchymal stem cells and the GLP-1 receptor agonist exenatide could protect against premature nephropathy in diabetic rats [19]. Xian et al. [20] validated the protective effect of umbilical cord mesenchymal stem cells combined with resveratrol on podocyte injury in NOD mice. The proposal of combined drug administration provides a new strategy for the treatment of DN. In addition, a clinical trial has shown that although improvements in the condition of patients with lupus nephritis were observed after injection of MSCs, some immunosuppressive drugs are still required for maintenance treatment [21]. Therefore, considering the future clinical applications of MSCs, combined therapy is also regarded as a worthwhile research topic. However, whether the combination of Irbesartan with MSCs can improve the treatment of DN through their respective therapeutic mechanisms remains to be further investigated.

MSCs, as "living" drugs, have pharmacokinetic parameters that are difficult to evaluate through conventional analytical methods, once they enter the body, these cells are akin to entering a black box. In recent years, radiolabeling and nuclear imaging tracking of adoptively transferred cells offer a promising noninvasive approach to studying their PK profile in vivo. Among them, it mainly includes positron emission tomography (PET) and single photon emission computed tomography (SPECT). In contrast, PET imaging has greater advantages, such as higher resolution, higher detection sensitivity, and less radioactivity [22]. However, a significant drawback of using nuclide labeling methods is the possibility of intracellular radioactive isotopes leaking out during cellular metabolism and cell death [23, 24]. This creates difficulties in distinguishing between live cells, damaged cells, radioactive cell fragments, or leaked nuclides, thereby impacting the precision of tracer results [25]. Recently, Yang et al. [26] proposed that radiolabeled cells can be tracked in vivo through positron emission tomography (PET) imaging to trace the trajectory of cell migration. They found that the use of the iron chelators deferoxamine (DFO) can enhance the accuracy of PET imaging, reduce bone uptake, preserve uptake in major organs, and avoid interference from free isotopes released by dead cells. Hence, we employ this method to investigate the in vivo trajectory of MSCs and confirm their ability to home to damaged kidney tissue, consequently exerting a protective effect on renal function.

Materials and methods

Isolation, culture and identification of umbilical cord-derived mesenchymal stem cells

UC-MSCs were obtained from Xuzhou Regional Cell Preparation Center Co., Ltd. Fresh umbilical cord tissue was collected and washed with phosphate-buffered saline (PBS), after which the Wharton's jelly was separated from the umbilical cord tissue using sterile scissors and forceps. The Wharton's jelly was then cut into small pieces approximately 1 mm3 in size. These pieces were cultured in 15 mL of serum-free medium in T75 flasks. Upon reaching approximately 80% confluence, the cells were passaged. We used fifth passage cells for subsequent experiments. Cells expressed CD73, CD90, and CD105 but did not express CD19, CD34, CD45, CD79a, CD11b, or HLA-DR. These cells met the current international standards for defining mesenchymal stem cells derived from bone marrow [27]. Flow cytometry results were provided by Nanjing Kingmed for Clinical Laboratory.

PET imaging of 89Zr labeled MSCs in DN model

To prepare the 89Zr-oxine complex, we obtained a certain volume of oxalic acid 89Zr solution, added 9 times the volume of HEPES buffer solution (0.1 mol/L) and added 0.375 times the volume of Na2CO3 solution (1 mol/L) to adjust the pH to 7. Then, we labeled mesenchymal stem cells with 89Zr-oxine. The cells were centrifuged and distributed into 15 mL sterile centrifuge tubes. Then, 10 μCi of labeled 89Zr-oxine was added to each 1 × 106 stem cell sample and incubated for 15 min at room temperature. The cells were intermittently agitated to ensure that the cells were suspended. Then, the mixture was centrifuged at 300×g for 5 min at room temperature and washed 3 times with DPBS to remove unbound 89Zr-oxine and free 89Zr ions to obtain 89Zr-stem cells (89Zr-MSCs). Finally, we intravenously injected 89Zr-labeled mesenchymal stem cells into the rats. To eliminate free Zirconium from the body, 200 μL of 50 mg/mL DFO was injected into the muscle of each rat 30 min before the injection of 89Zr-MSCs. At 1 h, 3 h, 6 h, 24 h, 48 h, 72 h, 5 d, 10 d, or 12 d after 89Zr-MSCs administration, an Inveon MicroPET scanner (Siemens Medical Solutions) was used for static PET scanning. PET images were quantitatively analyzed using a previously reported method [28].

Antibody arrays

Rat Cytokine arrays (GSR-CYT-3-1, RayBiotech, Norcross, GA, USA) were used according to the manufacturer’s instructions to measure the expression levels of 27 cytokines in in the renal cortex of rats. The slide scanning was performed using InnoSan 300 Microarray Scanner (Innopsys, Parc d'Activités Activestre; 31 390 Carbonne-France). Differentially expressed proteins were arranged using hierarchical clustering and represented as a heat map. Data analysis was conducted using the GSR-CYT-3 data analysis software.

Drugs and chemicals

Irbesartan (Sanofi-Aventis, France, 30 mg/kg/day, IG) and streptozotocin (STZ) (Sigma Chemical Co., St. Louis, MO, USA, 35 mg/kg, IP) were used. A Rat Cytokine Array GS3 was used (RayBiotech, USA). Nephrin (Santa Cruz, sc-376522), WT-1 (Abcam, ab267377), and NPHS2 (Proteintech, 20384-1-AP) were used. Serum creatinine (Scr) and blood urea nitrogen (BUN) levels were measured by a Hitachi automatic analyzer (Hitachi Co. Ltd., Tokyo, Japan). Urinary albumin was measured by standard methods using commercial ELISA kits (Wuhan Newqidi Biological Technology Co., Ltd.).

Animal

Animal experiments were approved by the Experimental Animal Ethics Committee of Xuzhou Medical University (202207s017). Forty male Sprague-Dawley (SD) rats weighing 160–180 g were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., and raised in the Animal Center of Xuzhou Medical University. The sample size of the experimental and control groups was determined based on previous studies regarding the treatment of DN using MSCs transplantation combined with other drugs [27, 29] and our pilot trial. The laboratory temperature was 22 °C, and an alternative 12-h light/dark cycle was used. There were 4–5 rats per cage. Ear tags were used for labeling. These rats had free access to food and water and could freely move in their cages.

Animal experiments

After the animals were acclimatized and fed for one week, they were randomly divided into the following five groups using a random number generator: (1) the control group (CON), (2) the diabetic nephropathy group (DN), (3) the mesenchymal stem cells treatment group (MSCs), (4) the irbesartan treatment group (Irb), and (5) the combined administration group (MSC + Irb). In order to control experimental bias, we requested a researcher unfamiliar with the animal modeling situation to perform blind grouping, with the number of rats that contributed data for analysis in each experiment indicated in figure legends. Rats in the control group were fed normal chow, and the remaining rats were fed high-fat chow (Research Diets, Shanghai Synergy; XTHF45 rodent diet with 45 kcal% fat) for 6 weeks. Except for control rats the rats were intraperitoneally injected with streptozotocin (35 mg/kg; Sigma-Aldrich, St. Louis, MO, USA) dissolved in 0.1 M sodium citrate (pH 4.5), and control rats were injected with 0.1 M sodium citrate [6]. Three days after STZ injection, blood glucose levels were measured by tail vein blood sampling for three consecutive days, and blood glucose levels above 16.7 mmol/L were considered to indicate diabetes [30]. Body weight was monitored weekly, and blood glucose levels were tested every two weeks. After 4 weeks of streptozotocin injection, the diabetic rats exhibited significant increases in urine output, and urine protein levels greater than 20 mg/24 h indicated successful modeling [17]. During the experiment, we tried to minimize the pain of the animals and did not experience any adverse events related to the experimental drug. After animal modeling, we closely observe the state of the experimental animals. Due to the serious progression of some animal diseases that meet humanitarian priorities and end of experiment indicators, such as anorexia and weakness, they should be removed from the experiment and euthanasia should be implemented in a timely manner. The rats in the MSCs group and the MSC + Irb group were injected with 5th generation UC-MSCs (2 × 106 cells, 1 mL) via the tail vein 3 times every 10 days [31]. The remaining groups were injected with an equal amount of PBS via the tail vein. Rats in the Irb group and MSC + Irb group were administered irbesartan (30 mg/kg/d) by gavage. The remaining groups received the same amount of saline by gavage. Ensure that each group has at least 6 rats.

For the pharmacokinetic experiment, animal breeding and modeling were similar to the pharmacological experiments, and the animals were divided into two groups. (1) the 89Zr-MSCs-CON (C + M) group (n = 5); (2) the 89Zr-MSCs-DN (D + M) group (n = 5). The meaning is respectively the injection of 89Zr-labeled mesenchymal stem cells in control group and diabetic nephropathy group. 10 SD rats in total.

Before dissection, rats were euthanized with an overdose of pentobarbital sodium. Our animal studies were reported in accordance with the ARRIVE guidelines 2.0. (Additional file 1).

Assessment of glucose tolerance

After 3 weeks of drug treatment, the rats were fasted for 12 h, and glucose solution (1 g/kg) was injected intraperitoneally. Blood glucose was measured by collecting blood from the tail vein at 0, 25, 30, 60, and 120 min after injection. Insulin resistance was assessed by the steady-state test (HOMA-IR) and was calculated using the following formula: HOMA-IR = glucose concentration (mmol/L) × insulin (μU/L)/22.5.

Renal histopathological evaluation

Kidney tissues were fixed in 10% formalin for 24 h, embedded in paraffin, sectioned at a thickness of 3 μm, stained with hematoxylin-eosin (HE) and periodic acid-Schiff (PAS), and examined under a microscope. To prepare ultrathin sections, 1 mm3 of kidney tissue was fixed with 2.5% glutaraldehyde (pH 7.4) for 24 h at 4 °C. The tissue was then fixed in 1% osmium tetroxide for 2–3 h, dehydrated in acetone and ethanol, and embedded in epoxy resin. Sections were cut to a thickness of 50 nm with an ultramicrotome and then stained with 3% uranyl acetate and lead citrate. The sections were examined by transmission electron microscopy.

Urine and blood samples and tissue specimens

Urine samples were collected from the rats before the end of the experiment, and to prevent the effects of feces and food on urinary proteins, the rats were placed in metabolic cages (Shanghai, China) and were fasted but watered. Urine was collected and centrifuged at 1500 rpm for 5 min, and the supernatant was collected and stored at −80 °C. These samples were used to measure urinary microalbumin and creatinine. The urine microalbumin/creatinine ratio (UACR) was subsequently calculated. Before the end of the experiment, the rats were fasted for 12 h and anesthetized with sodium pentobarbital for blood sampling. The collected blood was centrifuged at 3000×g for 15 min, and the supernatant was stored at −80 °C. These samples were used to measure blood creatinine, blood urea nitrogen, and inflammatory factor levels. Left kidney tissue was fixed in 10% formalin for histological examination. The other part of the renal cortex was cut into 1 mm × 1 mm × 1 mm cubes and placed in electron microscopy fluid for storage (stored at 4 ℃ in the dark). The right cortex was quickly frozen in liquid nitrogen and stored at −80 °C for biochemical and immunoblot analyses.

Western blotting

Kidney tissue was added to lysis buffer (Bi Biotechnology, Shanghai, China), cut into pieces with scissors and processed with a grinder. Total protein was separated by SDS-PAGE and transferred to an NC membrane (Millipore, MA, USA). The membranes were blocked with protein-free rapid blocking solution for 15 min and rapidly washed with 1× PBS for 2 min. The NC membranes were incubated overnight at 4 ℃ with the following antibodies: anti-nephrin (1:1000; Santa Cruz; sc-376522), anti-NPHS2 (1:1000; Proteintech; 20384-1-AP), anti-WT-1 (1:1000; Abcam; ab267377), and β-actin (1:10000; Bioworld; AP0060). Then, the membranes were incubated with secondary antibodies at room temperature. After 1 h, the protein bands were examined using near-infrared fluorescence imaging technology. The band intensity was analyzed using ImageJ software (NIH, Bethesda, MD, USA).

Quantitative reverse transcription PCR (real-time PCR)

Total RNA was extracted from approximately 30 mg of kidney tissue using TRIzol® reagent. The RNA concentration was measured by a super-trace ultraviolet spectrophotometer (Thermo). Total RNA (1 μg) was reverse transcribed. The following primer pairs were used: GAPDH forward 5′-CTGGAGAAACCTGCCAAGTATG-3′, reverse 5′-GGTGGAAGAATGGGAGTTGCT-3′; β-NGF forward 5′-GAGACTCTGTCCCTGAAGCCCA-3′, reverse 5′-CCACAGTGATGTTGCGGGTCT-3′; GM-CSF forward 5′-TACAGTTTCTCAGCACCCACC-3′, reverse 5′-TCTTCGTTCTTTTCGTTCTCCAG-3′; TNF-α forward 5′-CCAGGTTCTCTTCAAGGGACAA-3′, reverse 5′-GGTATGAAATGGCAAATCGGCT-3′; and IL-1β forward 5′-TGTGACTCGTGGGATGATGAC-3′, reverse 5′-CCACTTGTTGGCTTATGTTCTGTC-3′. Relative mRNA expression was normalized to the GAPDH level and calculated by the 2−ΔΔCT method.

Statistical analysis

Statistical analysis was performed using SPSS statistics 20.0 software and GraphPad Prism 9.0 (GraphPad Software, Inc., San Diego, CA, USA). All data are presented as the mean ± SEM. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Differences between two groups were assessed with Student’s t test. P < 0.05 indicated statistical significance.

Results

Expression of cell surface markers to identify UC-MSCs

P1 UC-MSCs were analyzed using flow cytometry to identify cell surface markers. Results showed positive expression of CD105 (99.94%), CD90 (99.97%), and CD73 (100.00%) (Fig. 1A–C). However, CD34, CD45, CD11b, CD19, CD79a, and HLA-DR were all negatively expressed, with respective negative rates of 0.31%, 0.2%, 0.19%, 0.12%, 0.59%, and 0.00% (Fig. 1D–H). These findings indicate that UC-MSCs exhibit high expression of mesenchymal stem cell markers while lacking expression of hematopoietic stem cell markers, consistent with the international definition of MSCs.

Fig. 1
figure 1

The surface markers of UC-MSCs. A–I The surface markers were identified by flow cytometry. CD105, CD90 and CD73 were expressed, and the positive rates were 94%, 99.97% and 100.00%, respectively. CD34, CD45, CD11b, CD19 and CD79a were not expressed, and the expression rates were 0.31%, 0.2%, 0.19%, 0.12% and 0.59%, respectively. HLA-DR was not expressed, and the expression rate was 0.00%

Effects of UC-MSCs and irbesartan alone or in combination on the basic parameters of type 2 diabetic nephropathy

The control group had normal diet and water intake, were in good mental condition and were active. T2DN rats exhibited obvious polydipsia, polyphagia, polyuria and weight loss. The hair was yellow, the mental state was poor, and the movements were slow.

We measured body weight (Fig. 2A), fasting blood glucose (Fig. 2B), the HOMA-IR (Fig. 2C) and IPGTT (Fig. 2D–E). The body weights of DN rats were significantly lower than those of the control group. There was no notable change in body weight in any of the treatment groups. Compared to those in the control group, fasting blood glucose levels in the DN groups were significantly increased. However, UC-MSCs significantly decreased these levels; and combination treatment also induced decreases, but the difference was not significant. In addition, we examined glucose tolerance and insulin resistance indices in the rats. DN rats had impaired glucose metabolism and insulin resistance, and these conditions were significantly improved in the MSC + Irb group.

Fig. 2
figure 2

Effects of UC-MSC, irbesartan or the combination treatment on the biochemical indices of DN rats. A Body weight. B Fasting blood glucose. C HOMA-IR index. D IPGTT. E The AUC of the IPGTT. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Data were shown as mean ± SEM, n = 6. *P < 0.01 and ****P < 0.0001 versus the CON group, #P < 0.05 versus the DN group. IPGTT: Intraperitoneal glucose tolerance test, AUC: area under the curve, HOMA-IR: homeostasis model assessment index for insulin resistance, CON: control group, DN: diabetes nephropathy group, MSCs: mesenchymal stem cells treatment group, Irb: irbesartan treatment group, MSC + Irb: combined administration group

Effects of UC-MSCs and irbesartan alone or in combination on the renal function parameters of type 2 diabetic nephropathy

To evaluate the reno-protective effects of UC-MSCs and irbesartan alone or in combination on diabetic rats, we measured the urinary microalbumin/creatinine ratio (UACR), kidney index, and serum creatinine (Scr) and blood urea nitrogen (BUN) levels in each group. After treatment, the UACR was significantly lower in all the groups than in the DN group, and the effect was most significant in the combination administration group (Fig. 3A). The kidney indices in the DN group were significantly higher than those in the control group. The kidney indices were decreased in all the treatment groups but were only significantly different in the combined administration group (Fig. 3B). Serum creatinine and blood urea nitrogen (Fig. 3C–D) levels were higher in DN rats than in control rats. Serum creatinine levels were significantly lower in all the groups after treatment, but this difference was significant only in the combined administration group. Blood urea nitrogen was significantly reduced in the UC-MSC treatment group and combined administration treatment group, but in the irbesartan group, there was a reduction but not a significant difference. More importantly, the effects were most significant in the combined administration group.

Fig. 3
figure 3

Effects of UC-MSC, irbesartan or the combination treatment on renal parameters in DN rats. A Urine albumin creatine ratio. B Kidney indices. C Serum creatine levels. D Blood urea nitrogen levels. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Data were shown as mean ± SEM, n = 6. **P < 0.001, ***P < 0.001 and ****P < 0.0001 versus the CON group, #P < 0.05, ##P < 0.01 and ###P < 0.001 versus the DN group. CON: control group, DN: diabetes nephropathy group, MSCs: mesenchymal stem cells treatment group, Irb: irbesartan treatment group. MSC + Irb: combined administration group

Effects of UC-MSCs and irbesartan alone or in combination on the renal histopathology

We performed HE (Fig. 4A) and PAS (Fig. 4B) staining and found that, compared with those in the control group, renal pathological changes in diabetic rats were obvious and mainly characterized by increased glomerular volume, mesangial cell proliferation, an increase in the extracellular matrix, and thickening of the glomerular basement membrane. After treatment in each group, these pathological changes were eliminated.

TEM (Fig. 4C) showed that DN rats had significant fusion of foot processes and thickening of the glomerular basement membrane, and these effects were improved after the treatment of each group.

Fig. 4
figure 4

Histopathological analysis of the tissues of DN rats. A Representative images of H&E-stained kidney tissues in the different groups (scale bars = 100 μm). B Representative images of periodic acid–Schiff staining of kidney tissues in the different groups (scale bars = 100 μm), arrowhead indicate expansion of the mesangial matrix. C Representative transmission electron microscopy images of kidney tissues in the different groups (scale bar = 500 nm), arrowhead represents foot processes of the podocytes; asterisk represents GBM. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Data were shown as mean ± SEM, n = 6. ****P < 0.0001 versus the CON group, #P < 0.05, ##P < 0.01 and ####P < 0.0001 versus the DN group. CON: control group, DN: diabetes nephropathy group, MSCs: mesenchymal stem cells treatment group, Irb: irbesartan treatment group, MSC + Irb: combined administration group

Effects of UC-MSCs and irbesartan alone or in combination against podocyte injury in rats

WB experiments showed that the expression of the podocyte injury marker proteins podocin and nephrin was significantly lower in the DN group than in the control group (Fig. 5A–B). The three groups showed significant improvements after treatment. However, the improvement in the combined administration group was the most evident. In addition, the expression of the podocyte nuclear protein WT-1 (Fig. 5C) was significantly reduced in the DN group, indicating that rats in the DN group experienced significant podocyte loss. Podocyte loss in DN rats in each group improved after treatment, and the therapeutic effect in the combined administration group was the most significant. Uncropped images can be found in Figures S2–4.

Fig. 5
figure 5

The effects of UC-MSCs, irbesartan, and the combination treatment on podocyte injury. A–C Representative Western blots showing podocin, nephrin and WT-1 in the renal cortex. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Data were shown as mean ± SEM, n = 6. ***P < 0.001 and ****P < 0.0001 versus the CON group, #P < 0.05, ##P < 0.01, ###P < 0.001 and ####P < 0.0001 versus the DN group. CON: control group, DN: diabetes nephropathy group, MSCs: mesenchymal stem cells treatment group, Irb: irbesartan treatment group, MSC + Irb: combined administration group. The immunoblot has been cropped, full-length blots are presented in Supplementary Figs. 2–4

Safety and homing effect of UC-MSCs

In our study, 89Zr-labeled mesenchymal stem cells were injected into rats through the tail vein, and a microPET device was used for imaging.

Using microPET imaging technology, we examined the distribution of mesenchymal stem cells in the body at the start of the experiment (Fig. 6D). The results showed that within a short period of time, the injected clinical-grade mesenchymal stem cells were concentrated mainly in the lungs. PET imaging revealed no obvious signs of pulmonary embolism, which provided preliminary evidence of the safety of our mesenchymal stem cell therapy.

Consistent with the findings of previous studies, most bone marrow MSCs were distributed in the lungs and liver and had limited engraftment capacity [32]. The uptake in the lung, liver and kidney is shown in Fig. 6A–C. In addition, PET imaging showed that UC-MSCs underwent renal homing at 3 h after injection (Fig. 6D). This effect suggested that these cells could migrate to the damaged area, which is important for the treatment of various diseases.

These observations provide important insights into the safety and efficacy of mesenchymal stem cell therapy in rat models of normal and diabetic nephropathy, providing a basis for further research and potential clinical applications.

Fig. 6
figure 6

Quantitative analysis of UC-MSCs in different tissues and PET imaging. A–C Quantitative analysis of radiolabeled MSCs in the lungs, liver, and kidneys of the different groups of rats. D Representative PET images of each treatment group from Day 0 to Day 12. C + M: Injection of 89Zr-MSCs in the control group. D + M: Injection of 89Zr-MSCs in the diabetic nephropathy group

Effects of UC-MSCs and irbesartan alone or in combination on the expression of renal inflammatory factors in rats

We used an inflammatory factor antibody chip to examine inflammatory factors in the kidney cortex. A total of 27 inflammatory factors were identified on the chip. Cluster analysis (Fig. 7A) showed that the enrichment of TNF-α, IL-1β, β-NGF, GM-CSF and IL-6 in the kidney tissues of the DN group was higher than that in the CON group. The concentrations of TNF-α, IL-1β, β-NGF and GM-CSF were decreased after treatment with UC-MSCs alone or combined with irbesartan, but irbesartan alone had no significant effects on these inflammatory factors (Fig. 7B–D). In addition, the results of qPCR also confirm the above conclusion (Fig. 7E–H). The enrichment of IL-2 in the kidney tissues of the DN group was lower than that in the CON group, and it decreased further after treatment with irbesartan alone or in combination (Fig. 7B, D). These results suggest that UC-MSCs and irbesartan may synergistically enhance the anti-inflammatory effect of the combined treatment group by regulating different inflammatory factors.

Fig. 7
figure 7

The effects of UC-MSC, irbesartan, and the combination treatment on the expression of inflammatory factors. A–D Cytokines that were differentially expressed are shown as a heat map (red represents high content, and blue represents low content). D–G The expression of TNF-α, IL-1β, β-NGF, and GM-CSF was determined by qPCR. Comparisons among groups were performed using one-way analysis of variance (ANOVA). Data were shown as mean ± SEM, n = 6. *P < 0.05, **P < 0.01 and ****P < 0.0001 versus the CON group, #P < 0.05, ##P < 0.01 and ####P < 0.0001 versus the DN group. CON: control group, DN: diabetes nephropathy group, MSCs: mesenchymal stem cells treatment group, Irb: irbesartan treatment group, MSC + Irb: combined administration group

Discussion

This study is the first to demonstrate the reno-protective effects of irbesartan combined with UC-MSCs. These effects may be attributed to the modulation of various inflammatory factors within the rat kidneys by MSCs and irbesartan, synergistically ameliorating podocyte injury and thereby improving diabetic nephropathy. Moreover, this paper was the first to demonstrate the homing of MSCs to the kidneys of rats with diabetic nephropathy using micro-PET.

Diabetic nephropathy (DN) is a common microvascular complication of diabetes mellitus and a leading cause of end-stage renal disease [33]. Although its pathogenesis is unclear, diabetes induces adaptive pathological changes in the glomerulus, such as thickening of the glomerular basement membrane (GBM), the tethered matrix, and glomerular capillaries [34]. Moreover, with the development of DN, the structure and function of glomerular podocytes are markedly altered; for example, there is skeletal rearrangement in podocytes, podocyte fusion or even disappearance, an increase in the formation of intercellular tight junctions, a decrease in the length of slit septa, and a decrease in the number of podocytes [35]. This dysfunction leads to changes in intraglomerular capillary permeability, damage to the glomerular filtration barrier (GFB) and ultimately proteinuria [36]. These pathological changes can be monitored by measuring changes in the levels of nephrin, podocin and WT-1. Specifically, nephrin and podocin play key roles in regulating and stabilizing the integrity of the glomerular filtration barrier and renal function; WT-1 is expressed in the nucleus of podocytes, and its expression indirectly reflects the number of podocytes in the glomerulus [37]. Nephrin, podocin and WT-1 play important roles in maintaining the integrity of GFB, and decreases in their expression can result in podocyte apoptosis, detachment and fusion, ultimately leading to symptoms of renal insufficiency, such as proteinuria. Overall, glomerular podocyte injury is a key step in the progression of DN, and ameliorating podocyte loss and increasing the expression of podocyte-associated proteins, such as nephrin and podocin, can significantly slow the progression of proteinuria and protect the kidney. In addition, inflammation is a crucial pathological characteristic observed in chronic kidney diseases [38], including DN.

At present, there are various treatment methods for DN, including anti hyperglycemic drugs, lipid-lowering drugs, and renin angiotensin aldosterone system inhibitors (RAASi) [39]. The most widely used among them is RAASi, which includes angiotensin converting enzyme inhibitors (ACEIs) and angiotensin receptor inhibitors (ARBs) [40]. Among them, irbesartan has been shown to exhibit dose-dependent renoprotective effects on diabetic patients [41], as well as superior renal prognostic effects than the other drugs used in the study (amlodipine, placebos and antihypertensive drugs) [42]. In addition, compared with losartan and valsartan, irbesartan has higher bioavailability, lower plasma protein binding and a longer half-life. It has also been proven to reduce oxidative stress in diabetic kidneys [43]. Tunçdemir et al. [44] found that irbesartan can regulate kidney hemodynamics and reduce podocyte injury, thus exhibiting renoprotective effects on animal kidneys. Additionally, irbesartan can also reduce the production of proteinuria [45] and possesses a certain anti-inflammatory effect [46].

Recently, an increasing number of animal studies and clinical trials have highlighted the significant therapeutic potential of MSCs for DN [12, 47,48,49]. Previous studies have reported that MSCs have the potential to promote renal regeneration through their anti-inflammatory and anti-fibrotic effects [50, 51]. Li et al. [52] revealed that early administration of MSCs could maintain the balance of the immune microenvironment through an immunoregulatory mechanism, which prevented the occurrence of kidney injury, renal dysfunction, and glomerulosclerosis. Moreover, MSCs significantly reduce the expression of proinflammatory factors, including IL-1β, IL-6, and TNF-α, in the kidneys of DN rats [53]. The regulation of inflammation and tissue repair is related to the ability of MSCs to home to the site of inflammation to repair damaged tissue and maintain tissue homeostasis [54]. Because UC-MSCs contain younger stem cell populations, it has been suggested that they have greater regenerative potential. UC-MSCs also have the advantages of increased culture stability, increased replication potential, decreased immunogenicity, and increased accessibility; thus, many studies have focused on UC-MSCs [55].

In addition, a previous study showed that the combination of MSCs with the GLP-1 receptor agonist exenatide exerted significant protective effects against early-onset nephropathy in diabetic rats [27]. Other researchers have shown that the combination of UC-MSCs with the plant compound resveratrol protected against inflammation-induced glomerular podocyte injury in NOD mice [56]. The combination of cellular drugs with traditional therapeutic agents provides new ideas for the treatment of DN.

The FBG and UACR of the rats demonstrated the successful establishment of the DN model. We confirmed that the combination of MSCs and irbesartan led to a significant reduction in UACR, kidney index, BUN levels, serum creatinine levels, IPGTT and other relevant renal function parameters in rats with diabetic nephropathy. Furthermore, the combination therapy group showed significant improvements in parameters such as IPGTT, kidney index, and serum creatinine levels in T2DN rats, which were not observed in either the mesenchymal stem cell treatment group or the irbesartan treatment group. Interestingly, during the experiments, fasting blood glucose levels were greatly reduced in DN rats that were treated with UC-MSCs but were reduced but not significantly different in the rats that received the combination treatment. We believe that this effect is mainly due to interindividual differences. Differences in the dose and number of injections notably affected the hypoglycemic effect [20]. Previous studies have suggested that this hypoglycemic effect occurs because MSCs stimulate pancreatic β-cell regeneration [57].

Given the clear mechanism by which irbesartan treats diabetic nephropathy, primarily involving the regulation of the renin-angiotensin-aldosterone system (RAAS), our study focused on UC-MSCs. As living cell therapeutics, the pharmacological effects and pharmacokinetic characteristics of UC-MSCs are closely linked. Therefore, we believe that monitoring the in vivo biological distribution and behavior of injected cells is crucial. To validate the distribution of UC-MSCs in the bodies of DN rats, we administered 89Zr-labeled UC-MSCs and utilized PET imaging technology for visualization. Consistent with prior studies, our observations indicate that MSCs survive in the bodies of rats for approximately 10 days, primarily homing to the lungs and liver initially, followed by migration to other organs, including the kidneys [33, 58, 59]. Additionally, we enhanced the accuracy and reliability of our imaging results by administering intramuscular DFO injections to eliminate false-positive results caused by free nuclides.

In addition, we examined the expression of inflammatory factors in rat kidney tissues by using an inflammatory factor antibody chip. We found that the enrichment of TNF-α, IL-1β, β-NGF, GM-CSF and IL-6 in the kidney tissues of the DN group was higher than that in the CON group. The expression levels of TNF-α, IL-1β, β-NGF and GM-CSF were significantly decreased in the MSC alone and combination groups, but no significant difference was observed in the irbesartan group. Compared with the CON group, the expression of IL-2 in the renal cortex of DN decreased, while it increased after treatment with irbesartan and combination therapy, with no significant difference observed in the MSCs treatment group. Among them, β-NGF is a member of the nerve growth factor family that regulates the development and function of the nervous system in combination with other nerve growth factors. An increase in NGF expression under high glucose conditions can lead to renal diseases such as diabetic nephropathy [29]. Neuroprotective growth factors such as NGF, GDNF and BDNF play important roles in podocyte differentiation, apoptotic resistance and cytoskeletal protection. Granulocyte-macrophage colony-stimulating factor (GM-CSF) [60] plays an important role in regulating and promoting the growth, differentiation and function of white blood cells in the hematopoietic system. These results suggest that UC-MSCs and irbesartan may have a synergistic anti-inflammatory effect through the regulation of different cytokines when administered in combination.However, there are certain limitations in the research process of this article, previous research only explored the pharmacological effects of combined administration without investigating issues such as different drug dosages or treatment durations. Combining our pharmacokinetic experimental results, we will explore and validate these issues in subsequent experiments, aiming to further elucidate the mechanism of combined administration. Furthermore, it is essential to consider the influence of animal gender factors in future design of animal experiments.

In conclusion, the combination of umbilical cord-derived mesenchymal stem cells and irbesartan may be a promising therapeutic strategy for diabetic nephropathy and provides a new idea for the clinical translation of MSCs. Due to the complexity of DN pathogenesis, we will focus on the specific pathways in future studies.

Conclusion

In summary, we found that the combined application of umbilical cord-derived mesenchymal stem cells and irbesartan significantly improved glomerular podocyte damage in rats with diabetic nephropathy. Additionally, we observed that mesenchymal stem cells homed to the injured kidneys and regulated the expression of inflammatory factors. This may the potential mechanism by which mesenchymal stem cells exert their therapeutic effects. Therefore, these two treatments, which act through different mechanisms, synergistically protect the kidneys of rats with diabetic nephropathy. These findings provide new insights for the clinical application of mesenchymal stem cells.

Availability of data and materials

All remaining data and materials are available from the authors upon reasonable request. This protocol was not registered.

Abbreviations

BUN:

Blood urea nitrogen

DM:

Diabetes mellitus

DN:

Diabetic nephropathy

ELISA:

Enzyme Linked immunosorbent assay

ESRD:

End-stage renal disease

FBG:

Fasting blood glucose

GBM:

Glomerular basement membrane

GFB:

Glomerular filtration barrier

HE:

Hematoxylin and eosin staining

IHC:

Immunohistochemical

MSC:

Mesenchymal stem cell

PAS:

Periodic acid-Schiff stain

PBS:

Phosphate buffer saline

PET:

Positron emission tomography

qRT-PCR:

Quantificational real-time chain polymerase chain reaction

RAAS:

Renin-angiotensin-aldosterone system

SCr:

Serum creatinine

SEM:

Standard error of mean

TEM:

Transmission electron microscope

TG:

Triglyceride

UACR:

Urine albumin-to-creatinine ratio

WB:

Western Bolt

References

  1. Saagoo MK, Luigi G. Diabetic nephropathy: an overview. Methods Mol Biol (Clifton, NJ). 2020;2067:3–7. https://doi.org/10.1007/978-1-4939-9841-8_1.

    Article  CAS  Google Scholar 

  2. Muskiet MHA, et al. GLP-1 and the kidney: from physiology to pharmacology and outcomes in diabetes Nature reviews. Nephrology. 2017;13(10):605–28. https://doi.org/10.1038/nrneph.2017.123.

    Article  CAS  PubMed  Google Scholar 

  3. Webster AC, et al. Chronic kidney disease. Lancet. 2017;389(10075):1238–52. https://doi.org/10.1016/S0140-6736(16)32064-5.

    Article  PubMed  Google Scholar 

  4. Michal H-E, et al. Dedifferentiation of immortalized human podocytes in response to transforming growth factor-β: a model for diabetic podocytopathy. Diabetes. 2011;60(6):1779–88. https://doi.org/10.2337/db10-1110.

    Article  CAS  Google Scholar 

  5. Yasuno K, et al. Early-onset podocyte injury and glomerular sclerosis in Osborne–Mendel rats. J Vet Med Sci. 2010;72(10):1319–27. https://doi.org/10.1292/jvms.10-0094.

    Article  PubMed  Google Scholar 

  6. Ziyadeh F, Gunter W. Pathogenesis of the podocytopathy and proteinuria in diabetic glomerulopathy. Curr Diabetes Rev. 2008;4(1):39–45. https://doi.org/10.2174/157339908783502370.

    Article  CAS  PubMed  Google Scholar 

  7. State Key Laboratory of Dampness Syndrome of Chinese Medicine, T.S.A.H.o.G.U.o.C.M., Guangzhou, China, et al. Differences in serum proteins in traditional Chinese medicine constitutional population: analysis and verification. J Leukocyte Biol. 2020. 108(2):547–57. https://doi.org/10.1002/jlb.6vma0220-663rr

  8. Janaina P, et al. Mesenchymal stem cells as therapeutic candidates for halting the progression of diabetic nephropathy. Stem Cells Int. 2016;2016:1. https://doi.org/10.1155/2016/9521629.

    Article  CAS  Google Scholar 

  9. Park JH, et al. Delayed treatment with human umbilical cord blood-derived stem cells attenuates diabetic renal injury. Transpl Proc. 2012;44(4):1123–6. https://doi.org/10.1016/j.transproceed.2012.03.044.

    Article  CAS  Google Scholar 

  10. Imasawa T, et al. The potential of bone marrow-derived cells to differentiate to glomerular mesangial cells. J Am Soc Nephrol: JASN. 2001;12(7):1401–9. https://doi.org/10.1681/asn.v1271401.

    Article  PubMed  Google Scholar 

  11. Lauren B, et al. Potentiating renal regeneration using mesenchymal stem cells. Transplantation. 2019;103(2):307–13. https://doi.org/10.1097/tp.0000000000002455.

    Article  Google Scholar 

  12. Dongwei L, et al. Concise review: current trends on applications of stem cells in diabetic nephropathy. Cell Death Dis. 2020;11(11):1000–1000. https://doi.org/10.1038/s41419-020-03206-1.

    Article  Google Scholar 

  13. Neil R, et al. Cord blood in regenerative medicine: do we need immune suppression? J Transl Med. 2007;5(1):8. https://doi.org/10.1186/1479-5876-5-8.

    Article  CAS  Google Scholar 

  14. Hirano A, et al. Assessment of safety and feasibility of human allogeneic adipose-derived mesenchymal stem cells in a pediatric patient. Pediatric Res. 2018;84(5):575–7. https://doi.org/10.1038/s41390-018-0042-8.

    Article  Google Scholar 

  15. Yang C-C, et al. Repeated administration of adipose-derived mesenchymal stem cells added on beneficial effects of empagliflozin on protecting renal function in diabetic kidney disease rat. Biomed J. 2023;47(2):100613–100613. https://doi.org/10.1016/j.bj.2023.100613.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chen K-H, et al. Dose-dependent benefits of iron-magnetic nanoparticle-coated human umbilical-derived mesenchymal stem cell treatment in rat intracranial hemorrhage model. Stem Cell Res Ther. 2022. https://doi.org/10.1186/s13287-022-02939-4.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Ping N, et al. Human umbilical cord mesenchymal stem cells reduce oxidative damage and apoptosis in diabetic nephropathy by activating Nrf2. Stem Cell Res Ther. 2021;12(1):450–450. https://doi.org/10.1186/s13287-021-02447-x.

    Article  CAS  Google Scholar 

  18. Satirapoj B, Adler SG. Comprehensive approach to diabetic nephropathy. Kidney Res Clin Pract. 2014;33(3):121–31. https://doi.org/10.1016/j.krcp.2014.08.001.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lewis EJ, Hunsicker LG, Clarkeet WR, et al. Renoprotective effects of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. ACC Curr J Rev. 2002;11(1):26–26. https://doi.org/10.1016/S1062-1458(01)00550-5.

    Article  Google Scholar 

  20. Department of Endocrinology, T.A.H.o.Q.U., Qingdao 266003, China, et al. Protective effect of umbilical cord mesenchymal stem cells combined with resveratrol against renal podocyte damage in NOD mice. Diabetes Res Clin Pract. 2019. 156:107755. https://doi.org/10.1016/j.diabres.2019.05.034

  21. Habiba UE, et al. The therapeutic effect of mesenchymal stem cells in diabetic kidney disease. J Mol Med (Berl). 2024;102(4):537–70. https://doi.org/10.1007/s00109-024-02432-w.

    Article  PubMed  Google Scholar 

  22. Wei W, et al. Noninvasive PET imaging of T cells. Trends Cancer. 2018;4(5):359–73. https://doi.org/10.1016/j.trecan.2018.03.009.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Lechermann LM, et al. Detection limit of 89Zr-labeled T cells for cellular tracking: an in vitro imaging approach using clinical PET/CT and PET/MRI. EJNMMI Res. 2020;10(1):82. https://doi.org/10.1186/s13550-020-00667-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Man F, et al. In vivo PET tracking of 89Zr-Labeled Vγ9Vδ2 T cells to mouse xenograft breast tumors activated with liposomal alendronate. Mol Ther. 2019;27(1):219–29. https://doi.org/10.1016/j.ymthe.2018.10.006.

    Article  CAS  PubMed  Google Scholar 

  25. Gawne PJ, et al. Direct cell radiolabeling for in vivo cell tracking with PET and SPECT imaging. Chem Rev. 2022;122(11):10266. https://doi.org/10.1021/acs.chemrev.1c00767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xu Q, et al. Repurposing iron chelators for accurate positron emission tomography imaging tracking of radiometal-labeled cell transplants. MedComm. 2024;5(2):e473–e473. https://doi.org/10.1002/mco2.473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Habib HA, Heeba GH, Khalifa MMA. Effect of combined therapy of mesenchymal stem cells with GLP-1 receptor agonist, exenatide, on early-onset nephropathy induced in diabetic rats. Eur J Pharmacol. 2020;892:173721.

    Article  PubMed  Google Scholar 

  28. Yi W, et al. The multi-therapeutic role of MSCs in diabetic nephropathy. Front Endocrinol. 2021;12:671566–671566. https://doi.org/10.3389/FENDO.2021.671566.

    Article  Google Scholar 

  29. Jianxia H, et al. Effect of combined therapy of human Wharton’s jelly-derived mesenchymal stem cells from umbilical cord with sitagliptin in type 2 diabetic rats. Endocrine. 2014;45(2):279–87. https://doi.org/10.1007/s12020-013-9984-0.

    Article  CAS  Google Scholar 

  30. Jingjing L, et al. Quantitative radio-thin-layer chromatography and positron emission tomography studies for measuring streptavidin transduced chimeric antigen receptor T cells. J Chromatogr B. 2021;1182:122944–122944. https://doi.org/10.1016/J.JCHROMB.2021.122944.

    Article  Google Scholar 

  31. Department of Nephropathy, T.S.H.o.J.U., 218 Ziqiang Street, Changchun 130041, China, et al. Nrf2 participates in the anti-apoptotic role of zinc in Type 2 diabetic nephropathy through Wnt/β-catenin signaling pathway. J Nutr Biochem. 2020. 84(prepublish):108451–108451. https://doi.org/10.1016/j.jnutbio.2020.108451

  32. Lv S-S, et al. Mesenchymal stem cells transplantation ameliorates glomerular injury in streptozotocin-induced diabetic nephropathy in rats via inhibiting macrophage infiltration. Int Immunopharmacol. 2013;17(2):275–82. https://doi.org/10.1016/j.intimp.2013.05.031.

    Article  CAS  PubMed  Google Scholar 

  33. Creane M, et al. Biodistribution and retention of locally administered human mesenchymal stromal cells: quantitative polymerase chain reaction based detection of human DNA in murine organs. Cytotherapy. 2017;19(3):384–94. https://doi.org/10.1016/j.jcyt.2016.12.003.

    Article  CAS  PubMed  Google Scholar 

  34. Jing C, et al. TangShenWeiNing formula prevents diabetic nephropathy by protecting podocytes through the SIRT1/HIF-1α pathway. Front Endocrinol. 2022;13:888611–888611. https://doi.org/10.3389/FENDO.2022.888611.

    Article  Google Scholar 

  35. Yan W, et al. Mesenchymal stem cells: an overview of their potential in cell-based therapy for diabetic nephropathy. Stem Cells Int. 2021;2021:6620811–6620811. https://doi.org/10.1155/2021/6620811.

    Article  Google Scholar 

  36. Thomas MC, et al. Diabetic kidney disease. Nat Rev Dis Primers. 2015;1(1):15070. https://doi.org/10.1038/nrdp.2015.70.

    Article  Google Scholar 

  37. State Key Laboratory of Organ Failure Research, N.C.R.C.o.K.D., Nanfang Hospital, Southern Medical University, Guangzhou, China, et al. Diabetic kidney diseases revisited: a new perspective for a new era. Mol Metab. 2019. 30:250–63. https://doi.org/10.1016/j.molmet.2019.10.005

  38. Hans-Joachim A, Liliana S. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol. 2014;25(7):1387–400. https://doi.org/10.1681/asn.2014010117.

    Article  Google Scholar 

  39. Xinyue L, et al. Intravenous injection of human umbilical cord-derived mesenchymal stem cells ameliorates not only blood glucose but also nephrotic complication of diabetic rats through autophagy-mediated anti-senescent mechanism. Stem Cell Res Ther. 2023;14(1):146–146. https://doi.org/10.1186/s13287-023-03354-z.

    Article  CAS  Google Scholar 

  40. Hui-Teng C, et al. Gamma-secretase activity is dispensable for mesenchyme-to-epithelium transition but required for podocyte and proximal tubule formation in developing mouse kidney. Development (Cambridge, England). 2003;130(20):5031–42. https://doi.org/10.1242/dev.00697.

    Article  CAS  Google Scholar 

  41. Brenner BM, Cooper ME, de Zeeuw D. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. ACC Curr J Rev. 2002;11(1):26–26. https://doi.org/10.1016/S1062-1458(01)00549-9.

    Article  Google Scholar 

  42. Parving H-H, et al. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. New Engl J Med. 2001;345(12):870–8. https://doi.org/10.1056/NEJMoa011489.

    Article  CAS  PubMed  Google Scholar 

  43. Xiong X-L, et al. Irbesartan attenuates contrast media-induced NRK-52E cells apoptosis. Pharmacol Res. 2006;54(4):253–60. https://doi.org/10.1016/j.phrs.2006.05.005.

    Article  CAS  PubMed  Google Scholar 

  44. Tunçdemir M, Öztürk M. The effects of angiotensin-II receptor blockers on podocyte damage and glomerular apoptosis in a rat model of experimental streptozotocin-induced diabetic nephropathy. Acta Histochem. 2010;113(8):826–32. https://doi.org/10.1016/j.acthis.2010.12.003.

    Article  CAS  Google Scholar 

  45. Fang L-N, et al. Protective effect of irbesartan by inhibiting ANGPTL2 expression in diabetic kidney disease. Curr Med Sci. 2020;40:1–7. https://doi.org/10.1007/s11596-020-2304-z.

    Article  CAS  Google Scholar 

  46. Chen Y, et al. Effects of different doses of Irbesartan combined with spironolactone on urinary albumin excretion rate in elderly patients with early type 2 diabetic nephropathy. Am J Med Sci. 2018;355(5):418–24. https://doi.org/10.1016/j.amjms.2018.01.017.

    Article  PubMed  Google Scholar 

  47. Manizheh K, et al. Mesenchymal stem cell-based therapies against podocyte damage in diabetic nephropathy. Tissue Eng Regener Med. 2017;14(3):201–10. https://doi.org/10.1007/s13770-017-0026-5.

    Article  CAS  Google Scholar 

  48. Yuling H, Lina Y. Mesenchymal stem cells and extracellular vesicles in therapy against kidney diseases. Stem Cell Res Ther. 2021;12(1):219–219. https://doi.org/10.1186/s13287-021-02289-7.

    Article  Google Scholar 

  49. Bo L, Lilach O. MSC therapy for diabetic kidney disease and nephrotic syndrome. Nature reviews. Nephrology. 2023;19(12):754–5. https://doi.org/10.1038/s41581-023-00776-z.

    Article  Google Scholar 

  50. Xiang E, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Therapy. 2020;11(1):336. https://doi.org/10.1186/s13287-020-01852-y.

    Article  CAS  Google Scholar 

  51. Christoph K, Rafael K. Role of mesenchymal stem cells in kidney injury and fibrosis. Curr Opin Nephrol Hypertens. 2016;25(4):372–7. https://doi.org/10.1097/mnh.0000000000000230.

    Article  Google Scholar 

  52. Agudelo JSH, et al. Mesenchymal stromal cell-derived microvesicles regulate an internal pro-inflammatory program in activated macrophages. Front Immunol. 2017;8:881. https://doi.org/10.3389/fimmu.2017.00881.

    Article  CAS  Google Scholar 

  53. Lewis EJ, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. New Engl J Med. 2001;345(12):851–60. https://doi.org/10.1056/NEJMoa011303.

    Article  CAS  PubMed  Google Scholar 

  54. Yuanmin L, et al. Early intervention with mesenchymal stem cells prevents nephropathy in diabetic rats by ameliorating the inflammatory microenvironment. Int J Mol Med. 2018;41(5):2629–39. https://doi.org/10.3892/ijmm.2018.3501.

    Article  CAS  Google Scholar 

  55. Hanieh KA, et al. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: the homing procedure and their effective parameters. Eur J Haematol. 2023;112(2):153–73. https://doi.org/10.1111/EJH.14014.

    Article  Google Scholar 

  56. Tan L, et al. Human umbilical cord mesenchymal stem cells: an overview of their potential in cell-based therapy. Expert Opin Biol Ther. 2015;15(9):1293–306. https://doi.org/10.1517/14712598.2015.1051528.

    Article  CAS  Google Scholar 

  57. Hamza AA, et al. Mechanistic insights into the augmented effect of bone marrow mesenchymal stem cells and thiazolidinediones in streptozotocin-nicotinamide induced diabetic rats. Sci Rep. 2018;8(1):1–18. https://doi.org/10.1038/s41598-018-28029-1.

    Article  CAS  Google Scholar 

  58. Jingjing H, et al. Amelioration of diabetic nephropathy in mice by a single intravenous injection of human mesenchymal stromal cells at early and later disease stages is associated with restoration of autophagy. Stem Cell Res Ther. 2024;15(1):66–66. https://doi.org/10.1186/s13287-024-03647-x.

    Article  CAS  Google Scholar 

  59. Gayong S, et al. Pharmacokinetics and in vivo fate of intra-articularly transplanted human bone marrow-derived clonal mesenchymal stem cells. Stem cells Develop. 2015;24(9):1124–32. https://doi.org/10.1089/scd.2014.0240.

    Article  CAS  Google Scholar 

  60. Perlman AS, et al. Serum inflammatory and immune mediators are elevated in early stage diabetic nephropathy. Ann Clin Lab Sci. 2015;45(3):256–63.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Xuzhou Regional Cell Preparation Center Co., Ltd, for providing an ample supply of umbilical cord-derived mesenchymal stem cells for our experiments.

Funding

This work was supported by the Natural Science Foundation of China [No. 82173883]; the Traditional Chinese Medicine Technology Development Plan Project of Jiangsu Province [No. MS2023175]; the Science and Technology Foundation of Xuzhou [No. KC22469]; the National College Students Innovation and Entrepteneurship Training Program; and the Science and Technology Planning Project of Jiangsu Province [No. BE2019636].

Author information

Authors and Affiliations

Authors

Contributions

Before conducting the experiment, we carefully designed the experimental plan and strictly followed it. XYZ, and XXY conceived and designed the study. JM, and XG collected and analyzed the data. JM, XG, XJL, ZW, YW, YHW, and ZQS performed the experiments. XYZ, and JM wrote the manuscript. All the authors read and approved the final manuscript. A protocol was prepared before the study.

Corresponding authors

Correspondence to Xiaoxing Yin or Xueyan Zhou.

Ethics declarations

Ethics approval and consent to participate

The animal study was conducted with the approval of and in accordance with the rules and regulations of the Institutional Animal Care and Use Committee of Xuzhou Medical University, Xuzhou, China (Title of the approved project: To explore the therapeutic effect of mesenchymal stem cells derived from human umbilical cord on diabetes nephropathy; approval number: 202207s017; date: 15 July 2022).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

13287_2024_3844_MOESM1_ESM.pptx

Additional file 1. Figure S1. Animal allocation situation. Figure S2. Full-length blots of podocin. Figure S3. Full-length blots of nephrin. Figure S4. Full-length blots of WT-1. Figure S5. The effect of Irbesartam on the cell viability, proliferation, aging, and apoptosis rate of UC-MSCs. CCK-8 assay; Cells were plated at equal density before challenged with Irbesartan (1 μM), the OD450 values of the CCK-8 test assay were detected after Irbesartan incubation for 0, 24, 48, 72 h respectively. (B) EdU labeling; Cells were exposed to EdU and then fixed and stained. Images were taken using fluorescence microscope (Olympus BX53), and cells labeled with EdU were Red. Scale bar = 100 μm. (C) Senesence associated β-galactosidase staining; The SA-β-gal-positive cells exhibited blue color (indicated by arrows) under phase-contact microscope. Scale bar = 50 μm. (D) Detection of apoptosis in MSCs by flow cytometry. Differences between two groups were assessed with Student’s t test. Data were shown as mean ± SEM, n = 6. MSCs: MSCs without irbesartan incubation, MSCs + Irb: MSCs incubated with Irbesartan. (PPTX 15962 kb)

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meng, J., Gao, X., Liu, X. et al. Effects of xenogeneic transplantation of umbilical cord-derived mesenchymal stem cells combined with irbesartan on renal podocyte damage in diabetic rats. Stem Cell Res Ther 15, 239 (2024). https://doi.org/10.1186/s13287-024-03844-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-024-03844-8

Keywords