HMGB1, TGF‑β and NF‑κB are associated with chronic allograft nephropathy

  • Authors:
    • Shi‑Qi Zhao
    • Zhen‑Zhen Xue
    • Ling‑Zhang Wang
  • View Affiliations

  • Published online on: October 17, 2017     https://doi.org/10.3892/etm.2017.5319
  • Pages: 6138-6146
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to investigate the association between high mobility group protein B1 (HMGB1), transforming growth factor‑β1 (TGF‑β1), nuclear factor‑κB (NF‑κB) and chronic allograft nephropathy (CAN) and to identify the clinical significance of HMGB1, TGF‑β1, NF‑κB on patients with CAN. Between September 2012 and November 2014, 27 patients with CAN diagnosed by biopsy were enrolled in the present study and a further 30 patients that underwent nephrectomy following trauma were selected as the control group. Immunohistochemical staining with HMGB1, TGF‑β1 and NF‑κB expression in the renal tissues, and western blot analysis were used to measure the relative expression of HMGB1, TGF‑β1 and NF‑κB. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was used to estimate the relative expression of HMGB1, TGF‑β1 and NF‑κB mRNA. Statistical analysis was used to calculate the association between HMGB1, TGF‑β1 and NF‑κB expression and CAN grade. Immunohistochemical staining demonstrated that HMGB1, TGF‑β1 and NF‑κB had markedly positive expression rates in renal tubular epithelial cell cytoplasm and membranes in CAN renal tissues, and the positive rates of HMGB1, TGF‑β1 and NF‑κB increased with the aggravation of CAN pathological grade (I, II and III). The results of western blot analysis indicated that the expression levels of HMGB1, TGF‑β1 and NF‑κB were significantly higher in the CAN group, compared with the normal group (P<0.05), and the expression levels increased with the progression of CAN grade. A positive association among HMGB1, TGF‑β1 and NF‑κB expression was identified. RT‑qPCR analysis demonstrated that the expression of HMGB1, TGF‑β1 and NF‑κB mRNA in the CAN group was significantly higher than in the normal group (P<0.05), and the relative expression level of HMGB1, TGF‑β1 and NF‑κB mRNA not only increased with the aggravation of CAN grade, but was also positively associated with the expression of HMGB1, TGF‑β1 and NF‑κB, respectively. The abnormal expression of HMGB1, TGF‑β1 and NF‑κB is therefore, an important manifestation of CAN and the expression of HMGB1, TGF‑β1 and NF‑κB mRNA in the renal tissues are significantly associated with CAN pathological progression. HMGB1, TGF‑β1 and NF‑κB may form a signaling pathway that leads to the occurrence of CAN, which induces renal interstitial fibrosis.

Introduction

Kidney transplantation has become the most efficient therapy for patients who suffer from end stage renal disease (1). However, despite the improvements of immunosuppressive methods on kidney transplantation, the long-term graft survival rate, such as the 5-year survival rate, has not improved markedly (2). Chronic allograft nephropathy (CAN) is one of the most common causes for loss of kidney graft function, which accounts for 50–80% of late graft loss after the first year of renal transplantation (3,4). The pathogenesis of CAN remains intricate and poorly understood, as the condition is caused by various immune and non-immune factors (5). For example, the immune factors include acute clinical rejection, human leukocyte antigen match, whereas non-immune factors consist of hypertension, glomerular hypertension, delayed graft function and acute calcineurin-inhibitor toxicity (68). These factors lead to cumulative damage to the kidney, which results in progressive allograft injury and consequently, dysfunction (9,10). A previous study also suggested that the effects of body mass index (BMI) 1 year after the kidney transplantation were associated with CAN (11). CAN is the process of fibrotic development with histological lesions, which can be characterized by atherosclerosis, glomerulopathy, interstitial fibrosis and tubular atrophy (6,9,12). Considering the negative effects that CAN causes on renal allograft, further insight on the pathology mechanisms is required and may provide potential therapeutics for CAN.

The high mobility group box 1 protein (HMGB1), which belongs to the most evolutionarily conserved proteins in the nucleus and cytoplasm, is one of the members of high mobility group nuclear protein family (13). HMGB1 has been demonstrated to act as a pro-inflammatory mediator and participate in human immune systems diseases (14,15). Studies indicated that innate immune cells, including macrophages and monocytes, actively secreted HMGB1 (13,16). It has been demonstrated that HMGB1 not only induces various cytokines, such as interleukin (IL)-1, IL-6 and IL-8, but also stimulates necrosis-induced inflammation. It indicates that HMGB1 has an important role in renal diseases, including acute kidney injury, chronic kidney disease (CKD) and granulomatous nephritis (1720). However, the association between HMBG1 and CAN remains to be clarified. Furthermore, HMGB1 acts as the endogenous ligand for toll-like receptor 2 (TLR2) and toll-like receptor 4 (TLR4), and is associated with downstream translocation of nuclear factor-κB (NF-κB) (21,22). NF-κB is a protein complex, which has an important role in transcription, immune responses and inflammation developments (23,24). The NF-κB family is a family of inducible transcription factors that have been demonstrated to contribute to the process of renal inflammation and immunological disease development (25,26). Transforming growth factor β (TGF-β) is a multi-function cytokine complex with three isoforms, TGF-β1, TGF-β2, and TGF-β3 (27). Among them, TGF-β1 has an important role in interstitial fibrosis development (2830). Multiple studies have suggested that the NF-κB pathway interacts with the TGF-β1/Smad pathway to form the NF-κB/TGF-β1/Smad signaling pathway, which is involved in the process of inflammation and renal tubulointerstitial fibrosis development (3133). Furthermore, the association between TGF-β1 expression and CAN has been identified in animal models and patients, demonstrating that there is an overexpression of TGF-β1 in patients with CAN (3,27). However, to the best of our knowledge, the role of NF-κB in CAN has not been investigated previously.

Therefore, the present study aimed to investigate whether HMGB1 and NF-κB in renal tissues are associated with CAN. Due to the association between TGF-β1 and CAN, TGF-β1 was included in the present study to investigate the relationship of TGF-β1 with HMGB1 and NF-κB in CAN. Ultimately, the current case-control study was conducted to accumulate data and determine the association between HMGB1, NF-κB, TGF-β1 and CAN.

Materials and methods

Subjects

A total of 27 patients (age range, 18–54 years old; mean age, 41±8 years; 15 males and 12 females) who were diagnosed with CAN by histological biopsy diagnosis between September 2012 and November 2014 at Linyi People's Hospital (Linyi, China) were enrolled into the study. All patients suffered graft kidney dysfunction between 1 and 10 years after allograft renal transplantation and the mean allograft survival time was 4.3 years. Every CAN patient received renal biopsy guided by B ultrasound to exclude other renal diseases and renal tissue specimens were harvested from each CAN patient. In addition, 30 cases (age range, 29–63 years old; mean age, 44±9 years; 16 males and 14 females) who received nephrectomy following trauma in the same time period were selected as the control group for the current study. Normal renal tissue specimens were collected through nephrectomy in the control group. Non-anticoagulant blood was collected under fasting conditions 1 day prior to renal biopsy to detect a number of physical indications, including hemoglobin, high sensitivity C-reactive protein (hs-CRP), IL-6, triglyceride (TG), low density lipoprotein (LDL-C), highdensity lipoprotein (HDL-C) and fasting plasma glucose (FPG). The present study was approved by the Ethics and Clinical Committee of Linyi People's Hospital and written informed consent was obtained from each patient.

Immunosuppressive regimens

Methylprednisolone (Pfizer Manufacturing Belgium, Puurs, Belgium; imported drug registration number: H20080285) was administered at a dose of 500 mg/day once a day by intravenous injection in the first 3 days following renal transplant. Subsequently, mycophenolate mofetil (Hangzhou Zhongmei Huadong Pharmaceutical Co. Ltd, Zhejiang Sheng, China; permission number approved by the state: H20052083) at 1.0–1.5 g/day was administered orally 48 h after the surgery. The concentration of serum creatinine (Cr) was assessed once a month using an automated biochemical analyzer (AU5800; Beckman Coulter, Inc., Brea, CA, USA). Cyclosporine A (CsA; Hangzhou Zhongmei Huadong Pharmaceutical Co. Ltd; permission number: H10960122) was administered at 8 mg/kg/day when the concentration of Cr was <250 µmol/l; and then adjusted to maintain the drug level at 300–350 ng/ml in the first 2 weeks following surgery, 250–300 ng/ml in weeks 2–4, 200–250 ng/ml in weeks 4–12 and 150–200 ng/ml in weeks 12–48 following surgery. Finally, prednisone (Zhejiang Xianju Pharmaceutical Co. Ltd., Zhejiang, China; permission number: H33021207) was used instead of methylprednisolones at 0.6 mg/kg/day and the dose was adjusted to maintain the blood drug concentration at 10–15 mg/day at 2 months after renal transplantation.

Classification of chronic allograft nephropathy

The diagnostic criteria and classification for all the specimens was assured according to the Banff 07 working classification (34). The classifications based on histopathological findings were as follows: Grade I (mild), mild interstitial fibrosis and tubular atrophy; Grade II (moderate), moderate interstitial fibrosis and tubular atrophy; and Grade III (severe), severe interstitial fibrosis and tubular atrophy and moderate tubular loss.

Histomorphology

For microscopic observation, tissues were fixed in 10% formalin at 37°C for 24 h and conventionally dehydrated (70% ethanol for 1 day, 80% ethanol for 45 min, 90% ethanol for 1 h, 95% ethanol for 1 h, and ethanol 100% for 3 h). Tissues were embedded in paraffin and 2-µm tissue sections were stained with hematoxylin and eosin (HE). For electron microscopy observation, renal tissues were fixed in 2.5% glutaraldehyde at 4°C for 4 h, and then fixed in 2% osmic acid (Seebio Biotech Co., Ltd., Shanghai, China) at 4°C for 2 h. Tissues were then dehydrated by a graded acetone series. Epon812 (Sangon Biotech Co., Ltd., Shanghai, China) was used for saturating and embedding tissues, following the manufacturer's protocol. An Ultra Rapid Tissue Processor (Histra-QS; Jokoh Co., Ltd., Tokyo, Japan) was used to produce 2-µm sections and uranyl acetate and lead citrate were applied to stain the tissues. A H-7500 transmission electron microscopy (magnification, ×7,000) was used to observe and capture images of the tissue sections.

Immunohistochemistry

Expression of HMGB1, TGF-β1 and NF-κB in renal tissue was detected by immunohistochemical staining (streptavidin-perosidase). Polyclonal antibodies against HMGB1 (#BA2361; 1:200), TGF-β1 (#BM4876; 1:200) and NF-κB p65 (#BM3946; 1:200) were purchased from Wuhan Boster Biological Engineering Co., Ltd., (Wuhan, China) The procedure was performed following the manufacturer's protocol of the VECTASTAIN® ABC kit (#PK-6100; Vector Laboratories, Inc., Burlingame, CA, USA). Tissues were fixed in 10% formalin at 37°C for 24 h and conventionally dehydrated (70% ethanol for 1 day, 80% ethanol for 45 min, 90% ethanol for 1 h, 95% ethanol for 1 h, and ethanol 100% for 3 h). Tissues were embedded in paraffin and 3-µm tissue sections were prepared. Following deparaffinization and rehydration to block endogenous peroxidase, sections were soaked in 0.003 (volume ratio) H2O2 in 80% methanol and washed with phosphate-buffered saline (PBS) three times. Normal non-immune serum was used to prevent unspecific binding. Subsequently, the primary antibody was added to sections overnight at 4°C, followed by overlaying with appropriate secondary antibody (#BM3895; 1:1,000; Wuhan Boster Biological Engineering Co., Ltd.) for 15 min at 37°C. PBS was used as the negative control instead of primary and secondary antibodies. Following 3,3′-diaminobenzidine (DAB) staining and hematoxylin counterstaining, a section of renal tissue was fixed with ethanol (75% for 1 min, 85% for 1 min, 95% for 1 min and 100% for 4 min). A light microscope (magnification, ×100) was used for observation and 10 high-power fields were randomly selected in each section to record the number of dying cells and calculate the positive signal rate. If there were no positive cells in each of the high power field on average, a ‘−’ grade was given. Positive cell numbers of <25%, 25–50% and >50% were graded as ‘+’, ‘++’ and ‘+++’, respectively.

Western blot analysis

Western blot analysis was used to detect the protein expression levels of HMGB1, TGF-β1 and NF-κB. The procedure was performed with the EasySee Western Blot kit (#DW101; Shanghai Bogoo Biotechnology Co., Ltd., Shanghai, China) according to the manufacturer's protocol. Polyclonal antibody rabbit anti-human HMGB1 (#BA2361; 1:200), TGF-β1 (#BM4876; 1:200) NF-κB p65 (#BM3946; 1:200), and goat anti-rabbit IgG conjugated to horseradish peroxidase (#BM3895; 1:1,000) were purchased from Wuhan Boster Biological Engineering Co., Ltd. Samples were treated with RIPA reagent (#P0013B; Beyotime Institute of Biotechnology, Shanghai, China) for 30 min at 4°C for protein extraction. A total of 10 µg cell plasma protein was separated by 10% SDS-PAGE. This was transferred onto a polyvinylidene fluoride membrane under a steady electric current (160 mA) for 20 min. The membrane was blocked with TBS-Tween-20 (TBST; Beyotime Institute of Biotechnology) and 5% skim milk at 37°C for 1 h. Then the samples were treated with primary antibodies at 4°C overnight. After washing with TBST 3 times, samples were treated with secondary antibodies at 37°C for 1 h. ChemiDocTM XRS gel imaging system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used for capturing images of the gel following DAB staining, and Image J software v1.48 (National Institutes of Health, Bethesda, MD, USA) was used to read integral absorbance values. Each sample was assessed with three replicates, and β-actin was used as an internal reference.

Reverse transcription-quantification polymerase chain reaction (RT-qPCR) analysis

RT-qPCR was used to detect the relative expression level of HMGB1, TGF-β1 and NF-κB mRNA relative to β-actin. Total RNA was isolated from renal tissues using an RNAprep Pure Tissue kit (#DP431; Tiangen Biotech Co., Ltd., Beijing, China), following the manufacturers protocol. In a 25-µl total reaction volume, 2 µg RNA was reverse transcribed into cDNA using an OligdT primer (Takara Bio, Inc., Otsu, Japan), following the protocol of 37°C for 15 min and 85°C for 5 sec. All PCR primers sequences are presented in the Table I. The full reaction components were: 0.4 µl PCR forward primer (10 µM), 0.4 µl PCR reverse primer (10 µM), 2 µl cDNA template, 7.2 µl RNase free dH2O and 10 µl SYBR Premix Ex Taq TM II (2×) (Takara Bio, Inc.). The amplification reaction was completed as follows: Pre-degeneration for 5 min at 95°C; degeneration for 1 min in 94°C; annealing for 60 sec at 59°C and extension for 60 sec at 72°C (35 cycles); and final extension for 8 min at 72°C. PCR product (5 µl) was used for electrophoresis on 1% agarose gel, and ethidium bromide (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for DNA stain. ChemiDocTM XRS gel imaging system (Bio-Rad Laboratories, Inc.) was used to image the agarose gel and Image-J software v1.48 was used to read its integral absorbance values. The relative expression level of mRNA was acquired by the ratio of relative intensity between the PCR products electrophoresis banding and the internal reference gene, β-actin.

Table I.

Primer sequences for quantitative polymerase chain reaction analysis.

Table I.

Primer sequences for quantitative polymerase chain reaction analysis.

GenePrimersProduct size (bp)
HMGB1Forward: 5′-TCATCTGCTGCAGTGTTGTT-3′285
Reverse: 5′-CTCAGAGAGGTGGAAGA-3′
TGF-β1Forward: 5′-TCCTGTGACAGCAGGGATAA-3′298
Reverse: 5′-TCCTGTGACAGCAGGGATAA-3′
NF-κBForward: 5′-CTGAACCAGGGCATACCTGT-3′197
Reverse: 5′-GAGAAGTCCATGTCCGCAAT-3′
β actinForward: 5′-AACCCTAAGGCCAACCGTGAAAAG-3′240
Reverse: 5′-TCATGAGGTAGTCTGTCAGGT-3′

[i] HMGB1, high mobility group protein B1; TGF-β1, transforming growth factor-β1; NF-κB, nuclear factor-κB; CAN, chronic allograft nephropathy; bp, base pairs.

Statistical analysis

SPSS 19.0 (IBM SPSS, Armonk, NY, USA) statistical software was used to analyze the data. Measurement data was expressed as mean ± standard deviation. The χ2 test was applied for enumeration data and Student's t-test and analysis of variance were applied for measurement data. Pearson correlation analysis was used to assess the protein expression level and mRNA relative expression level of HMGB1, TGF-β1 and NF-κB. Spearman correlation analysis was applied to analyze the association between CAN grade and HMGB1, TGF-β1 and NF-κB. A two-tailed P<0.05 was considered to indicate a statistically significant difference.

Results

Baseline characteristics

Clinical characteristics of CAN patients and controls are presented in Table II. No statistically significant differences were observed for age, sex, BMI index, hs-CRP, TG, LDL-C, HDL-C or FPG between CAN patients and controls (P>0.05). The glomerular filtration rate (GFR) and hemoglobin of CAN patients were significantly lower when compared with controls (P<0.05), whereas IL-6 was significantly higher in CAN patients than in controls (P<0.05; Table II).

Table II.

Clinical characteristics of CAN patients and controls.

Table II.

Clinical characteristics of CAN patients and controls.

CharacteristicCAN (n=27)Controls (n=30) χ2/tP-value
Age, years41±844±91.3240.19
Male/female15/1216/140.0280.87
GFR27±1087±2014.540<0.05
BMI24±426±41.8850.07
Hemoglobin, g/l132±15141±92.710<0.01
Hs-CRP, mmol/l4.7±1.24.3±1.21.2570.21
IL-6, ng/l27.4±8.515.1±4.76.659<0.05
TG, mmol/l2.1±1.41.9±0.90.6340.53
LDL-C, mmol/l2.9±1.32.6±1.10.9430.35
HDL-C, mmol/l1.3±0.61.4±0.50.6860.50
FPG, mmol/l5.5±0.55.3±0.61.3580.18

[i] Data are presented as the mean ± standard deviation. CAN, chronic allograft nephropathy; GFR, glomerular filtration rate; BMI, body mass index; Hs-CRP, high sensitivity C-reactive protein; IL-6, Interleukin-6; TG, triglyceride; LDL-C, low density lipoprotein; HDL-C, high density lipoprotein; FPG, fasting plasma glucose.

Histomorphology

In microscopic observations of paraffin sections with HE staining, no characteristic changes were observed, including abnormally elevated Cr induced by acute rejection or CsA toxicosis. However, interstitial fibrosis and tubular atrophy were observed based on the Banff 07 criteria, which were the primary characteristics of chronic allograft nephropathy. Among 27 renal tissues from CAN patients, there were 5 cases of CAN grade I, 12 cases of CAN grade II and 10 cases of CAN grade III. Observation results of light microscopy and electron microscopy are presented in Figs. 1 and 2, respectively.

Immunohistology

In the renal tissues of patients with CAN, HMGB1, TGF-β1 and NF-κB were markedly positively expressed in the cell cytoplasm and membrane of renal tubular epithelial cells (Fig. 3). HMGB1, TGF-β1 and NF-κB in the renal tissues of the CAN group revealed markedly higher positive rates than normal renal tissues (Table III). The positive rates of HMGB1, TGF-β1 and NF-κB in CAN grade III were markedly higher than in CAN grade I (Table III).

Table III.

Expression of HMGB1, TGF-β1 and NF-κB in the CAN and control groups.

Table III.

Expression of HMGB1, TGF-β1 and NF-κB in the CAN and control groups.

HMGB1TGF-β1NF-κB



GroupN++++++++++++++++++
Controls30191100181200181200
CAN I  5  0  221  0  221  0  122
CAN II12  0  246  0  264  0  273
CAN III10  0  136  0  127  0  046

[i] HMGB1, high mobility group protein B1; TGF-β1, transforming growth factor-β1; NF-κB, nuclear factor-κB; CAN, chronic allograft nephropathy; +, rate of positive cell number <25%; ++, rate of positive cell number 25–50%; +++, rate of positive cell number >50%.

Western blot analysis

As demonstrated by western blot analysis (Fig. 4A), the expression of HMGB1, TGF-β1, NF-κB was significantly higher in the renal tissues of patients in the CAN group, compared with normal tissues from the control group (P<0.05; Fig. 4B-D). Furthermore, statistically significant differences in the expression of HMGB1, TGF-β1 and NF-κB were identified among CAN grades I, II and III (P<0.05; Fig. 4). The expression of HMGB1 and NF-κB in the CAN grade I demonstrated no statistical difference when compared with the controls (P>0.05; Fig. 4B and C, respectively), whereas TGF-β1 expression was significantly higher compared with the control group (P<0.05; Fig. 4D). Furthermore, expression of HMGB1, NF-κB and TGF-β1 in CAN grade III was significantly higher than grade II (P<0.05; Fig. 4B-D, respectively).

RT-qPCR analysis

Compared with the control group, the relative expression of HMGB1, TGF-β1 and NF-κB mRNA in the CAN group was significantly elevated (P<0.05; Fig. 5A-C, respectively). Notably, there was significantly increased expression of HMGB1, TGF-β1 and NF-κB mRNA in CAN grade II, compared with grade I (P<0.05). Similarly, the expression of HMGB1, TGF-β1, NF-κB mRNA in CAN grade III were significantly higher compared with grade II (P<0.05; Fig. 5; Table IV).

Table IV.

Relative expression of HMGB1, TGF-β1 and NF-κB mRNA in the CAN and control groups.

Table IV.

Relative expression of HMGB1, TGF-β1 and NF-κB mRNA in the CAN and control groups.

CAN

CytokineCAN (n=27)Grade I (n=5)Grade II (n=12)Grade III (n=10)Controls (n=30)
HMGB1 0.85±0.13a 0.61±0.03b 0.86±0.02c0.96±0.040.41±0.03
TGF-β1 0.81±0.13a 0.57±0.03b 0.81±0.05c0.93±0.030.34±0.04
NF-kB 0.68±0.09a 0.56±0.04b 0.64±0.05c0.78±0.050.30±0.02

a P<0.05 vs. controls

b P<0.05 vs. CAN grade II

c P<0.05 vs. CAN grade III. Data are presented as the mean ± standard deviation. HMGB1, high mobility group protein B1; TGF-β1, transforming growth factor-β1; NF-κB, nuclear factor-κB; CAN, chronic allograft nephropathy.

Correlation analysis among HMGB1, TGF-β1 and NF-κB and CAN grade

Statistical analysis indicated positive associations between the expression levels of HMGB1, TGF-β1 and NF-κB in renal tissues (HMGB1 vs. TGF-β1: r=0.860, P<0.01; HMGB1 vs. NF-κB: r=0.899, P<0.01; TGF-β1 vs. NF-κB: r=0.835, P<0.01). In the renal tissues of patients with CAN, expression of HMGB1, TGF-β1 and NF-κB was positively associated with CAN pathological grade (HMGB1: r=0.894, P<0.01; TGF-β1: r=0.867, P<0.01; NF-κB: r=0.853, P<0.01) and the expression level of HMGB1, TGF-β1 and NF-κB tended to increase with the aggravation of the CAN pathological grade. Furthermore, a positive association between the protein and mRNA expression of HMGB1, TGF-β1 and NF-κB (r=0.904, P<0.01; r=0.858, P<0.01; r=0.885, P<0.01, respectively) was identified and the mRNA expression level of HMGB1, TGF-β1 and NF-κB in renal tissues also increased with the progression of the CAN pathological grade (Tables V and VI).

Table V.

Pearson correlation analysis of the association between the protein and mRNA expression levels of HMGB1, TGF-β1 and NF-κB (n=57).

Table V.

Pearson correlation analysis of the association between the protein and mRNA expression levels of HMGB1, TGF-β1 and NF-κB (n=57).

Analysis HMGB1TGFB1NF-κBHMGB1 mRNATGF-1 mRNANF-κB mRNA
HMGB1Pearson correlation10.8600.8990.904
P-value (2-tailed) <0.001<0.001<0.001
TGF-taiPearson correlation0.86010.8350.858
P-value (2-tailed)<0.001 <0.001 <0.001
NF-κBPearson correlation0.8990.83510.885
P-value (2-tailed)<0.001<0.001 <0.001
HMGB1 mRNAPearson correlation0.90410.9790.962
P-value (2-tailed)<0.001 <0.001<0.001
TGF-ailed)APearson correlation0.8580.97910.961
P-value (2-tailed) <0.001 <0.001 <0.001
NF-κB mRNAPearson correlation0.8850.9620.9611
P-value (2-tailed) <0.0010.000<0.001

[i] HMGB1, high mobility group protein B1; TGF-β1, transforming growth factor-β1; NF-κB, nuclear factor-κB.

Table VI.

Spearman correlation analysis of the association between the protein and mRNA expression levels of HMGB1, TGF-β1 and NF-κB and CAN grade (n=57).

Table VI.

Spearman correlation analysis of the association between the protein and mRNA expression levels of HMGB1, TGF-β1 and NF-κB and CAN grade (n=57).

VariableHMGB1TGFB1NF-κBHMGB1 mRNATGF-1 mRNANF-κB mRNA
Association0.8940.8670.8530.9150.9170.913
P-value (2-tailed)<0.001<0.001<0.001<0.001<0.001<0.001

[i] HMGB1, high mobility group protein B1; TGF-β1, transforming growth factor-β1; NF-κB, nuclear factor-κB; CAN, chronic allograft nephropathy.

Discussion

With the great developments made in transplantation immunology, the short-term graft survival rate has markedly improved, while the long-term survival rate has remained low (34). Studies have revealed that CAN is a leading factor, causing 50–80% of allograft loss in renal function in the late period following kidney transplantation (3,35). As CAN is caused by various immune and non-immune factors, and is easily affected by a series of donor-related factors including donor age, brain death and consequences of ischemia-reperfusion injury, the pathogenesis and mechanisms remain unclear (3640).

Previous studies suggested that HMGB1, which is a pro-inflammatory cytokine released from dying cells, would accumulate as renal function deteriorates and have an important role in CKD (17). The levels of HMGB1 may be markedly overexpressed during the progression of CKD. It was also demonstrated that HMGB1 functions are associated with NF-κB in the inflammatory and immunostimulation response (4143). NF-κB has also been implicated in kidney diseases and is associated with renal inflammatory injury and fibrosis (44). It is speculated that HMGB1-NF-κB may function as a signaling pathway in renal diseases. Previous studies revealed that TGF-β1 was regulated by NF-κB in certain signaling pathways, which indicates a potential role for NF-κB (45,46). Therefore, the present study aimed to investigate the association between HMGB1, NF-κB, TGF-β1 and CAN.

The current study analyzed the HMGB1, NF-κB and TGF-β1 staining intensity within graft kidneys. The results of immunohistological staining indicated that HMGB1, NF-κB and TGF-β1 were predominantly expressed in the mesangium and glomerular epithelium. The positive expression rate of HMGB1, NF-κB and TGF-β1 in CAN tissues was much higher when compared with controls, indicating potential associations between HMGB1, NF-κB, TGF-β1 and CAN. In the present study, the results of western blot analysis and RT-qPCR also demonstrated that HMGB1, NF-κB and TGF-β1 were significantly associated with CAN and specifically to the grade of CAN. HMGB1, NF-κB and TGF-β1 were significantly overexpressed in renal tissues of patients with CAN, compared with controls. The expression of HMGB1, NF-κB and TGF-β1 increased with the progression of CAN. Following the statistical analysis performed in the current study, a significant association between HMGB1, NF-κB and TGF-β1 in CAN progression was revealed.

TGF-β1 is one of the critical cytokines, which leads to renal fibrosis, causing renal interstitial fibrosis in end-stage renal disease (47). It has been proven to be involved in CAN pathogenesis (28,48,49). To the best of our knowledge, no studies have assessed the association between NF-κB and CAN. However, NF-κB was indicated to have a function in kidney diseases, including CKD (50). Furthermore, it has been demonstrated that NF-κB was associated with hepatic stellate cell in the apoptosis process, in which NF-κB activation stimulated TGF-β1, leading to serious renal interstitial fibrosis (5153). These findings suggested that NF-κB and TGF-β1 may constitute an important pathway in the pathogenesis of CAN, which may explain the positive association between NF-κB and TGF-β1 in the current study. HMGB1 has been studied in other renal diseases (54,55); however, it is another novel inflammatory marker, which, to the best of our knowledge, has not been studied in regard to CAN. The concentration of HMGB1 serum levels in CKD was elevated in a previous study (17). The overexpression of HMGB1 in tissues in the present study also demonstrated a marked association between HMGB1 and CAN. A potential explanation for the positive association between HMGB1 and NF-κB in the present study is that HMGB1 induces the downstream translocation of NF-κB by interacting with TLR2, TLR4 and RAGE (41,5658). TLR2, TLR, HMGB1/NF-κB/TGF-β1 may form a signaling pathway to induce the pathogenesis of CAN. The overexpression of HMGB1 stimulates NF-κB activation, leading to the overexpression of TGF-β1, which causes renal interstitial fibrosis and a series of immunostimulatory and inflammatory responses through the pathway, resulting in CAN. With the accumulation of these factors in renal tissues, the conditions of CAN progressively deteriorate. Therefore, these factors (HMGB1, NF-κB and TGF-β1) in the tissues of CAN Grade III would be higher than those observed in CAN Grade I. Although this assumption is based on the present results and other studies, more details regarding the function of HMGB1, NF-κB and TGF-β1 in CAN pathogenesis are required to validate these results in further studies.

In conclusion, the present study identified that the levels of HMGB1, NF-κB and TGF-β1 in renal tissues are significantly associated with CAN. Furthermore, expression of HMGB1, NF-κB and TGF-β1 increases with CAN progression. Finally, the positive associations among HMGB1, NF-κB and TGF-β1 indicate that the HMGB1/NF-κB/TGF-β1 pathway may be one of the leading causes of CAN. The present study therefore provides a novel way to understand the mechanisms of CAN. Novel targets, including HMGB1 and NF-κB, were revealed in relation to CAN, and the HMGB1/NF-κB/TGF-β1 pathway may be the foundation of a novel target treatment for CAN. However, further studies are required to validate the roles of HMGB1, NF-κB and TGF-β1 in CAN and to investigate the mechanism of action in CAN pathogenesis. The present study may lead to the identification of effective methods to treat CAN and elevate the long-term graft survival rate following kidney transplantation.

References

1 

Cui Y, Huang Q, Auman JT, Knight B, Jin X, Blanchard KT, Chou J, Jayadev S and Paules RS: Genomic-derived markers for early detection of calcineurin inhibitor immunosuppressant-mediated nephrotoxicity. Toxicol Sci. 124:23–34. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Lamb KE, Lodhi S and Meier-Kriesche HU: Long-term renal allograft survival in the United States: A critical reappraisal. Am J Transplant. 11:450–462. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Cassidy H, Slyne J, O'Kelly P, Traynor C, Conlon PJ, Johnston O, Slattery C, Ryan MP and McMorrow T: Urinary biomarkers of chronic allograft nephropathy. Proteomics Clin Appl. 9:574–585. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Weir MR and Wali RK: Minimizing the risk of chronic allograft nephropathy. Transplantation. 87 8 Suppl:S14–S18. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Birnbaum LM, Lipman M, Paraskevas S, Chaudhury P, Tchervenkov J, Baran D, Herrera-Gayol A and Cantarovich M: Management of chronic allograft nephropathy: A systematic review. Clin J Am Soc Nephrol. 4:860–865. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Lukenda V, Mikolasevic I, Racki S, Jelic I, Stimac D and Orlic L: Transient elastography: A new noninvasive diagnostic tool for assessment of chronic allograft nephropathy. Int Urol Nephrol. 46:1435–1440. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Arndt R, Schmidt S, Loddenkemper C, Grünbaum M, Zidek W, van der Giet M and Westhoff TH: Noninvasive evaluation of renal allograft fibrosis by transient elastography-a pilot study. Transpl Int. 23:871–877. 2010.PubMed/NCBI

8 

Sayin B, Karakayali H, Colak T, Sevmis S, Pehlivan S, Demirhan B and Haberal M: Conversion to sirolimus for chronic allograft nephropathy and calcineurin inhibitor toxicity and the adverse effects of sirolimus after conversion. Transplant Proc. 41:2789–2793. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Haas M: Chronic allograft nephropathy or interstitial fibrosis and tubular atrophy: What is in a name? Curr Opin Nephrol Hypertens. 23:245–250. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Del Bello A, Rostaing L, Congy-Jolivet N, Sallusto F, Gamé X and Kamar N: Kidney nephrectomy after allograft failure. Nephrol Ther. 9:189–194. 2013.(In French). View Article : Google Scholar : PubMed/NCBI

11 

Wang K and Liu QZ: Effect analysis of 1-year posttransplant body mass index on chronic allograft nephropathy in renal recipients. Transplant Proc. 43:2592–2595. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Johnston O, Cassidy H, O'Connell S, O'Riordan A, Gallagher W, Maguire PB, Wynne K, Cagney G, Ryan MP, Conlon PJ and McMorrow T: Identification of β2-microglobulin as a urinary biomarker for chronic allograft nephropathy using proteomic methods. Proteomics Clin Appl. 5:422–431. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Zhu P, Xie L, Ding HS, Gong Q, Yang J and Yang L: High mobility group box 1 and kidney diseases (Review). Int J Mol Med. 31:763–768. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Srinivasan M, Banerjee S, Palmer A, Zheng G, Chen A, Bosland MC, Kajdacsy-Balla A, Kalyanasundaram R and Munirathinam G: HMGB1 in hormone-related cancer: A potential therapeutic target. Horm Cancer. 5:127–139. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, et al: HMGB1 in health and disease. Mol Aspects Med. 40:1–116. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Gong Q, Zhong S, Wang L, Guo H, Xiang Y, Ichim TE, Wang CY, Chen S, Gong F and Chen G: Neutralization of the extracellular HMGB1 released by ischaemic damaged renal cells protects against renal ischaemia-reperfusion injury. Nephrol Dial Transplant. 26:469–478. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Bruchfeld A, Qureshi AR, Lindholm B, Barany P, Yang L, Stenvinkel P and Tracey KJ: High Mobility Group Box Protein-1 correlates with renal function in chronic kidney disease (CKD). Mol Med. 14:109–115. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Leelahavanichkul A, Huang Y, Hu X, Zhou H, Tsuji T, Chen R, Kopp JB, Schnermann J, Yuen PS and Star RA: Chronic kidney disease worsens sepsis and sepsis-induced acute kidney injury by releasing High Mobility Group Box Protein-1. Kidney Int. 80:1198–1211. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Zakiyanov O, Kriha V, Vachek J, Zima T, Tesar V and Kalousova M: Placental growth factor, pregnancy-associated plasma protein-A, soluble receptor for advanced glycation end products, extracellular newly identified receptor for receptor for advanced glycation end products binding protein and high mobility group box 1 levels in patients with acute kidney injury: A cross sectional study. BMC Nephrol. 14:2452013. View Article : Google Scholar : PubMed/NCBI

20 

Oyama Y, Hashiguchi T, Taniguchi N, Tancharoen S, Uchimura T, Biswas KK, Kawahara K, Nitanda T, Umekita Y, Lotz M and Maruyama I: High-mobility group box-1 protein promotes granulomatous nephritis in adenine-induced nephropathy. Lab Invest. 90:853–866. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Bhatelia K, Singh K and Singh R: TLRs: Linking inflammation and breast cancer. Cell Signal. 26:2350–2357. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Zhou TB: Role of high mobility group box 1 and its signaling pathways in renal diseases. J Recept Signal Transduct Res. 34:348–350. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Pateras I, Giaginis C, Tsigris C, Patsouris E and Theocharis S: NF-κB signaling at the crossroads of inflammation and atherogenesis: Searching for new therapeutic links. Expert Opin Ther Targets. 18:1089–1101. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Hayden MS and Ghosh S: Regulation of NF-κB by TNF family cytokines. Semin Immunol. 26:253–266. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Sanz AB, Sanchez-Niño MD, Ramos AM, Moreno JA, Santamaria B, Ruiz-Ortega M, Egido J and Ortiz A: NF-kappaB in renal inflammation. J Am Soc Nephrol. 21:1254–1262. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Mohammed-Ali Z, Cruz GL and Dickhout JG: Crosstalk between the unfolded protein response and NF-κB-mediated inflammation in the progression of chronic kidney disease. J Immunol Res. 2015:4285082015. View Article : Google Scholar : PubMed/NCBI

27 

Harris S, Coupes BM, Roberts SA, Roberts IS, Short CD and Brenchley PE: TGF-beta1 in chronic allograft nephropathy following renal transplantation. J Nephrol. 20:177–185. 2007.PubMed/NCBI

28 

Daniel C, Vogelbacher R, Stief A, Grigo C and Hugo C: Long-term gene therapy with thrombospondin 2 inhibits TGF-β activation, inflammation and angiogenesis in chronic allograft nephropathy. PLoS One. 8:e838462013. View Article : Google Scholar : PubMed/NCBI

29 

Ding Y and Choi ME: Regulation of autophagy by TGF-β: Emerging role in kidney fibrosis. Semin Nephrol. 34:62–71. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Choi ME, Ding Y and Kim SI: TGF-β signaling via TAK1 pathway: Role in kidney fibrosis. Semin Nephrol. 32:244–252. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Li Y, Ge Y, Liu FY, Peng YM, Sun L, Li J, Chen Q, Sun Y and Ye K: Norcantharidin, a protective therapeutic agent in renal tubulointerstitial fibrosis. Mol Cell Biochem. 361:79–83. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Ka SM, Yeh YC, Huang XR, Chao TK, Hung YJ, Yu CP, Lin TJ, Wu CC, Lan HY and Chen A: Kidney-targeting Smad7 gene transfer inhibits renal TGF-β/MAD homologue (SMAD) and nuclear factor κB (NF-κB) signalling pathways and improves diabetic nephropathy in mice. Diabetologia. 55:509–519. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Lan HY and Chung AC: TGF-β/Smad signaling in kidney disease. Semin Nephrol. 32:236–243. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Srinivas TR and Oppenheimer F: Identifying endpoints to predict the influence of immunosuppression on long-term kidney graft survival. Clin Transplant. 29:644–653. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Xia SQ, Fan Y, Tan MY and Zheng JH: Five-year follow-up after conversion from calcineurin inhibitor to sirolimus-based treatment in kidney transplant patients with chronic allograft nephropathy. Int J Clin Exp Med. 8:3552–3558. 2015.PubMed/NCBI

36 

Shrestha B and Haylor J: Experimental rat models of chronic allograft nephropathy: A review. Int J Nephrol Renovasc Dis. 7:315–322. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Timsit MO, Yuan X, Floerchinger B, Ge X and Tullius SG: Consequences of transplant quality on chronic allograft nephropathy. Kidney Int Suppl. S54–S58. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Leca N: Focal segmental glomerulosclerosis recurrence in the renal allograft. Adv Chronic Kidney Dis. 21:448–452. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Liao QB, Guo JQ, Zheng XY, Zhou ZF, Li H, Lai XY and Ye JF: Test performance of sputum microRNAs for lung cancer: A meta-analysis. Genet Test Mol Biomarkers. 18:562–567. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Schinstock CA, Stegall M and Cosio F: New insights regarding chronic antibody-mediated rejection and its progression to transplant glomerulopathy. Curr Opin Nephrol Hypertens. 23:611–618. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Tan Y, Wang Q, She Y, Bi X and Zhao B: Ketamine reduces LPS-induced HMGB1 via activation of the Nrf2/HO-1 pathway and NF-κB suppression. J Trauma Acute Care Surg. 78:784–792. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Shi Z, Lian A and Zhang F: Nuclear factor-κB activation inhibitor attenuates ischemia reperfusion injury and inhibits Hmgb1 expression. Inflamm Res. 63:919–925. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Zhou XJ, Dong ZG, Yang YM, Du LT, Zhang X and Wang CX: Limited diagnostic value of microRNAs for detecting colorectal cancer: A meta-analysis. Asian Pac J Cancer Prev. 14:4699–4704. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Terhzaz S, Overend G, Sebastian S, Dow JA and Davies SA: The D. Melanogaster capa-1 neuropeptide activates renal NF-kB signaling. Peptides. 53:218–224. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Kim HJ, Kim JG, Moon MY, Park SH and Park JB: IκB kinase γ/nuclear factor-κB-essential modulator (IKKγ/NEMO) facilitates RhoA GTPase activation, which, in turn, activates Rho-associated KINASE (ROCK) to phosphorylate IKKβ in response to transforming growth factor (TGF)-β1. J Biol Chem. 289:1429–1440. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Jia QQ, Wang JC, Long J, Zhao Y, Chen SJ, Zhai JD, Wei LB, Zhang Q, Chen Y and Long HB: Sesquiterpene lactones and their derivatives inhibit high glucose-induced NF-κB activation and MCP-1 and TGF-β1 expression in rat mesangial cells. Molecules. 18:13061–13077. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Oo YH, Shetty S and Adams DH: The role of chemokines in the recruitment of lymphocytes to the liver. Dig Dis. 28:31–44. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Saigo K, Akutsu N, Maruyama M, Otsuki K, Hasegawa M, Aoyama H, Matsumoto I, Asano T and Kenmochi T: Study of transforming growth factor-β1 gene, mRNA, and protein in Japanese renal transplant recipients. Transplant Proc. 46:372–375. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Assadiasl S, Ahmadpoor P, Nafar M, Pezeshki Lessan M, Pourrezagholi F, Parvin M, Shahlaee A, Sepanjnia A, Nicknam MH and Amirzargar A: Regulatory T cell subtypes and TGF-β1 gene expression in chronic allograft dysfunction. Iran J Immunol. 11:139–152. 2014.PubMed/NCBI

50 

Liu H, Sun W, Wan YG, Tu Y, Yu BY and Hu H: Regulatory mechanism of NF-kappaB signaling pathway on renal tissue inflammation in chronic kidney disease and interventional effect of traditional Chinese medicine. Zhongguo Zhong Yao Za Zhi. 38:4246–4251. 2013.(In Chinese). PubMed/NCBI

51 

Braz MM, Ramalho FS, Cardoso RL, Zucoloto S, Costa RS and Ramalho LN: Slight activation of nuclear factor kappa-B is associated with increased hepatic stellate cell apoptosis in human schistosomal fibrosis. Acta Trop. 113:66–71. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Guicciardi ME and Gores GJ: Apoptosis as a mechanism for liver disease progression. Semin Liver Dis. 30:402–410. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Qin L and Han YP: Epigenetic repression of matrix metalloproteinases in myofibroblastic hepatic stellate cells through histone deacetylases 4: Implication in tissue fibrosis. Am J Pathol. 177:1915–1928. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Liu GQ, Zuo XH, Jiang LN, Zhang YP, Zhang LM, Zhao ZG and Niu CY: Inhibitory effect of post-hemorrhagic shock mesenteric lymph drainage on the HMGB1 and RAGE in mouse kidney. Ren Fail. 38:131–136. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Qie GQ, Wang CT, Chu YF and Wang R: Expression of HMGB1/RAGE protein in renal carcinoma and its clinical significance. Int J Clin Exp Pathol. 8:6262–6268. 2015.PubMed/NCBI

56 

Karuppagounder V, Arumugam S, Thandavarayan RA, Pitchaimani V, Sreedhar R, Afrin R, Harima M, Suzuki H, Nomoto M, Miyashita S, et al: Modulation of HMGB1 translocation and RAGE/NFκB cascade by quercetin treatment mitigates atopic dermatitis in NC/Nga transgenic mice. Exp Dermatol. 24:418–423. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Kang N, Hai Y, Yang J, Liang F and Gao CJ: Hyperbaric oxygen intervention reduces secondary spinal cord injury in rats via regulation of HMGB1/TLR4/NF-κB signaling pathway. Int J Clin Exp Pathol. 8:1141–1153. 2015.PubMed/NCBI

58 

Sun J, Shi S, Wang Q, Yu K and Wang R: Continuous hemodiafiltration therapy reduces damage of multi-organs by ameliorating of HMGB1/TLR4/NFκB in a dog sepsis model. Int J Clin Exp Pathol. 8:1555–1564. 2015.PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 14 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao SQ, Xue ZZ and Wang LZ: HMGB1, TGF‑β and NF‑κB are associated with chronic allograft nephropathy. Exp Ther Med 14: 6138-6146, 2017
APA
Zhao, S., Xue, Z., & Wang, L. (2017). HMGB1, TGF‑β and NF‑κB are associated with chronic allograft nephropathy. Experimental and Therapeutic Medicine, 14, 6138-6146. https://doi.org/10.3892/etm.2017.5319
MLA
Zhao, S., Xue, Z., Wang, L."HMGB1, TGF‑β and NF‑κB are associated with chronic allograft nephropathy". Experimental and Therapeutic Medicine 14.6 (2017): 6138-6146.
Chicago
Zhao, S., Xue, Z., Wang, L."HMGB1, TGF‑β and NF‑κB are associated with chronic allograft nephropathy". Experimental and Therapeutic Medicine 14, no. 6 (2017): 6138-6146. https://doi.org/10.3892/etm.2017.5319