Korean J Physiol Pharmacol 2025; 29(1): 57-66
Published online January 1, 2025 https://doi.org/10.4196/kjpp.24.155
Copyright © Korean J Physiol Pharmacol.
Su-Jeong Choi1,#, Giang-Huong Vu1,#, Harsha Nagar1, Seonhee Kim1, Ikjun Lee1, Shuyu Piao1, Byeong Hwa Jeon1, Kaikobad Irani2, Sang-Ha Oh3,*, and Cuk-Seong Kim1,*
1Department of Physiology & Medical Science, College of Medicine, Chungnam National University, Daejeon 34134, Korea, 2Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA, 3Department of Plastic and Reconstructive Surgery, College of Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
Correspondence to:Sang-Ha Oh
E-mail: djplastic@cnu.ac.kr
Cuk-Seong Kim
E-mail: cskim@cnu.ac.kr
#These authors contributed equally to this work.
Author contributions: C.S.K. and S.H.O. designed this research. S.J.C., G.H.V., S.K., I.L., and S.P. performed data curation. S.J.C. conducted experiment. S.J.C. and G.H.V. wrote manuscript. H.N., B.H.J., K.I., S.H.O., and C.S.K. edited the manuscript. S.H.O. and C.S.K. supervised this research. S.J.C., B.H.J., K.I., S.H.O., and C.S.K. funded this research. All authors have read and agreed to the published version of the manuscript
Schwann cells are the most abundant cells in the peripheral nervous system, maintaining the development, function and regeneration of peripheral nerves. Defects in these Schwann cells injury response potentially contribute to the pathogenesis of diabetic peripheral neuropathy (DPN), a common complication of diabetes mellitus. The protein p66shc is essential in regulating oxidative stress responses, autophagy induction and cell survival, and is also vital in the development of DPN. In this study, we hypothesized that p66shc mediates high glucose-induced oxidative stress and autophagic dysfunction. In Schwann cells treated with high glucose; p66shc expression, levels of reactive oxygen species, autophagy impairment, and early apoptosis were elevated. Inhibition of p66shc gene expression by siRNA reversed high glucose-induced oxidative stress, autophagy impairment, and early apoptosis. We also demonstrated that the levels of p66shc was increased, while autophagy-related proteins p62 and LC3 (LC3-II/I) were suppressed in the sciatic nerve of streptozotocin-induced diabetes mice. P66shc-deficient mice exhibited the improvement in autophagy impairment after diabetes onset. Our findings suggest that the p66 plays a crucial role in Schwann cell dysfunction, identifying its potential as a therapeutic target.
Keywords: Autophagy, Diabetic peripheral neuropathy, Oxidative stress, p66shc, Schwann cells
Schwann cells (SCs) are the main glial cells of the peripheral nerve and are responsible for the development, function, and regeneration of peripheral nerves by ensheathment of unmyelinated axons, myelination of myelinated axons and secretion of neurotrophic factor [1]. In the process of peripheral nerve injury, SCs proliferate and phagocytize the disintegrated axons and myelin and then migrate to guide the regeneration direction of the newborn axon [2]. However, SCs are very sensitive to glucose and insulin levels and closely involved in the occurrence and development of type 1 diabetes mellitus, as well as diabetic peripheral neuropathy (DPN) which is a common diabetes’ complication [3]. The accumulating data demonstrated that the damage of SCs in diabetes mellitus caused neurotoxic intermediates accumulation and neurotrophic factors deficiency, contributing in axonal degeneration, finally leading to DPN [4,5]. However, the exact underlying mechanism regarding the involvement of SCs in the DPN pathogenesis is still not clear.
Autophagy is the process of continuously breaking down modified proteins or damaged organelles in lysosomes to maintain cell homeostasis and function [6]. It is characterized by the sequestering of organelles/aging or damaged proteins and the formation of autophagosomes, protecting cells from a variety of factors including oxidative stress and daily wear and tear [7]. Reactive oxygen species (ROS)-induced activation of PARP-1 promotes autophagy by activating AMP-activated protein kinase, likely
The ShcA family includes p46shc, p52shc, and p66shc; of these, p66shc are redox enzymes that regulates the level of intracellular ROS under cellular stress [17]. In diabetes, the increase in p66Shc levels have been reported in the peripheral blood mononuclear cells [18]. Moreover, numerous studies have shown that diabetic p66Shc-/- mice are protected from the onset of both micro- and macro-vascular complications compared to their wild-type counterpart [19-22]. It has been shown that p66Shc gene deletion increases the expression of antioxidant enzymes in endothelial cells, alleviates hyperglycemia-induced oxidative stress, and prevents diabetes-induced endothelial dysfunction in streptozotocin (STZ)-treated mice [22]. Besides, a hyperglycemia-induced increase in p66Shc expression was also found in human retinal endothelial cells and retinal pigment epithelial cells. Increased p66Shc expression resulted in functional damage and/or apoptosis in these cells, thus contributing to the development of diabetic retinopathy [23,24]. Interestingly, p66Shc has been reported to interact with LC3-II through the binding between a distinct N-terminal CH2 domain and three putative LC3 interacting region motifs, which is associated with p66shc-dependent autophagy [25]. The ability of p66shc to promote intracellular accumulation of ROS likely triggers metabolic imbalance and thus also autophagy.
We hypothesized that p66shc mediates high glucose-induced ROS generation and autophagy dysfunction. Therefore, we evaluated the role of SCs and related mechanisms in diabetic neuropathy.
Anti-Beclin1 antibody was purchased from Abgent. Anti-LC3-II and -LC3-II antibodies were from Novus Biologicals. Anti-p66shc was obtained from BD Biosciences. Anti-GRP78, anti-phospho-eIF2α, anti-total-eIF2α, and anti-CHOP were from Cell Signaling. Anti-p62 and the loading control anti-β actin were purchased from Sigma-Aldrich.
Rat SCs (S16) were obtained from the American Type Culture Collection and cultured in Dulbecco’s modified Eagle medium supplemented with 4 mM L-glutamine, 25 mM glucose, 1 mM sodium pyruvate, 1,500 mg/l sodium bicarbonate, 10% fetal bovine serum, 10 U/ml penicillin, and 10 µg/ml streptomycin (Lonza). The cells were incubated at 37°C in a 5% CO2 atmosphere. In duplicate, cultured cells were treated with 25, 50, 100, and 150 mM glucose for 72 h. S16 rat SCs were transfected with short interfering RNA (siRNA) for 72 h (rat p66shc siRNA: sense-UGAGUCUCUGUCAUCGCUGUU and antisense-CAGCGAUGACAGAGACUCAUU [Bioneer]) and negative control siRNA using the Lipofectamine 2000 reagent (Invitrogen) per the manufacturer’s recommendations.
The Institutional Animal Care and Use Committee of Chungnam National University (CNUH-019-A0058) approved the animal experiments. Six-week-old male C57BL/6 mice (Samtako, Inc.) were divided randomly into control and experimental groups. Diabetes was induced with STZ (2-deoxy-2-3-[methyl-3-nitrosoureido]-D-glucopyranose; Sigma) dissolved in 0.1 M citrate buffer (pH 4.5) and intraperitoneally injected at a dose of 50 mg/kg for 5 consecutive days. The control group was injected with citrate buffer vehicle. A glucometer (LifeScan) was used to measure blood glucose levels in tail venous blood every week. Following injection, the mice were monitored for symptoms of diabetes and body weight loss. The mice were weighed before STZ injection, and weekly thereafter, to monitor changes. All
S16 rat SCs were stained with 5 µM 2,7-dichlorodihydrofluo-rescein diacetate (H2-DCFDA) (Invitrogen) for 30 min at 37°C. Equal numbers of cells (10 × 104) were collected and transferred to a black 96-well plate. Fluorescence was analyzed using a fluorometer (Thermo Scientific) with a 485 nm excitation/530 nm emission filter.
The apoptosis of SCs was assessed by flow cytometry with Annexin V-FITC/propidium iodide (PI) staining according to the manufacturer’s instructions. S16 rat SCs were treated with the siRNA p66shc and glucose for 72 h, suspension and adherent cells were collected, washed twice with cold phosphate-buffered saline (PBS), and stained with Annexin V at room temperature for 15 min and PI at room temperature (apoptosis) and 5 µM H2-DCFDA for 30 min at 37°C (cellular ROS). The cells were analyzed by flow cytometry (BD Biosciences FACS). Annexin V-positive and PI-negative cells were considered early apoptotic and Annexin V-positive and PI-positive cells late apoptotic or dead. For ROS analysis, histogram shift was confirmed by measuring the glucose concentration or the flow cytometry value of S16 rat SCs treated with the siRNA p66shc.
For immunofluorescence staining, sciatic nerves were thoroughly washed with 0.01 M PBS, placed in sodium citrate, boiled, and cooled for 5 min. After washing three times with PBS, sections were placed in PBS with 10% goat serum and 0.1% Triton X-100 for 30 min. Samples were incubated overnight at 4°C with primary antibodies against mouse LC3 (1: 1,000), and p62 (1:500). After washing with PBS, the samples were incubated with Cy3 anti-rabbit or anti-mouse secondary antibody (1: 500) for 2 h at room temperature. Nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI). Sections were examined using a digital fluorescence microscope (DMI 6000; Leica Microsystems) equipped with a DAPI filter (365 nm). LC3, and p62 were visualized using a PE filter (546 nm) at 400× magnification, and images were captured with a photometric camera (Cool Snap HQ; Photometrics). S16 rat SCs were cultured on glass coverslips in 12-well plates in medium supplemented with 25 or 150 mM glucose for 72 h. After washing with PBS, cells were fixed with 4% (w/v) paraformaldehyde and permeabilized with 0.1% (v/v) Triton X-100. After blocking for 30 min with PBS containing 5% (w/v) bovine serum albumin and 5% (v/v) horse serum, cells were incubated overnight at 4°C with antibodies against LC3 (1:200), and p62 (1:200). The cells were labeled with a Alexa Fluor 488-conjugated secondary antibody (1:1,000) or Fluor 647-conjugated secondary antibody (1:1,000) for 1 h in the dark at room temperature. Images were obtained using a fluorescence microscope.
Sciatic nerve sections were homogenized in lysis buffer (20 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM ethylene glycol tetra acetic acid, 1% NP-40, 1% sodium deoxycholate, 2.5 mM sodium pyrophosphate, 1 mM sodium orthovanadate, 1 mM β-glycerophosphate, and a protease inhibitor cocktail) using a homogenizer (PRO200; PRO Scientific). Cells were harvested and the supernatants were collected. These were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis and transferred to a nitrocellulose membrane. After blocking with 5% non-fat dry milk in Tris-buffered saline containing 0.05% Tween 20, the membrane was incubated with the primary antibodies overnight at 4°C. The membrane was treated with an appropriate peroxidase-conjugated secondary antibody, and the chemiluminescent signal was developed using the SuperSignal West Pico or Femto substrate (Pierce Biotechnology). Band densities were quantified using the Gel Doc 2000 imaging system and Quantity One software (Bio-Rad). The membrane was reacted with an anti-β-actin antibody to normalize loading.
Chromosomal DNA was isolated from mouse tails (2–3 mm length). Polymerase chain reaction (PCR) was conducted using a KOD-FX (TOYOBO) genotyping kit according to the manufacturer’s instructions. The primer sequences were as follows: reverse (5'-TAA TAC GAC TCA CTA TAG GG-3') and forward (5'-AGG CGA TTA GTT GGG TA-3') for p66shc-RNAi KO. PCR conditions were 95°C for 5 min (one cycle), 95°C for 30 sec, 56°C for 45 sec, 72°C for 45 sec (33 cycles), and 72°C for 10 min.
Statistical analysis was performed using SPSS software (ver. 12.0 for Windows; SPSS Inc.). Student’s t-test and analysis of variance (ANOVA) with Tukey’s
As the importance of apoptosis in the induction of diabetes and the pathophysiology of different diabetes related complications have been reported in several papers [26-28], we evaluated apoptotic cell death of SCs in high glucose condition by performing Annexin V assay. As shown in Fig. 1A, FACS showed that high glucose (150 mM) in SCs significantly induced early apoptosis compared to the control (25 mM).
p66shc was reported as a pro-apoptotic protein that regulates oxidative stress and induces apoptosis by means of redox activity [29]. Since we observed high glucose-induced apoptosis in SCs, we investigated protein expression of p66shc. Our data showed a dose-dependent increase in protein expression of p66shc in SCs treated with high glucose (Fig. 1B). To further clarify whether the elevated p66shc level produced intracellular ROS causing apoptosis in SCs in high glucose conditions, we first inhibited p66shc protein expression with sip66shc. S16 cells were transfected with sip66shc, following with high glucose treatment for 72 h. p66shc deficiency was confirmed after siRNA transfection (Supplementary Fig. 1A). As expected, both accumulation of ROS and early apoptosis triggered by high glucose concentration was significantly abrogated by knockdown of p66shc (Fig. 1C, D). These results suggest that high glucose condition induces apoptosis
High glucose inhibits autophagy in chondrocytes and induces autophagy
p62 deletion impairs autophagosomes, exacerbating cell injury and misfolded protein stress in cardiomyocytes [32]. It was hypothesized that endoplasmic reticulum (ER) stress is activated in SCs treated with high glucose. We evaluated ER stress-related proteins. There was an overexpression of GRP78, p-eIF2α, CHOP in SC-treated high glucose (Supplementary Fig. 2A), and the p66shc deletion alleviated high glucose induced-ER stress (Supplementary Fig. 2B). These data collectively demonstrate that downregulation of p66shc recovers the high glucose-induced impairment of autophagy and ER stress in SCs.
p66shc expression was upregulated in SCs in high glucose condition. To confirm whether p66shc also showed increased expression in diabetic mice model, STZ-induced diabetic mice were employed. Six-week-old BL6 mice were consecutively injected with vehicle or 50 mg/kg/day STZ intraperitoneally for 5 days, and diabetes was observed for 8 weeks after onset. Diabetes in the mice was confirmed by monitoring their blood level and body weight before and after STZ injection. Two weeks after injection, the concentration of blood glucose in the diabetes group was higher than that of the control group; non-fasting blood glucose concentration of 250 mg/dl or more was defined as diabetes (Supplementary Fig. 3A). As the disease progressed, the body weight of STZ-treated mice did not change while that of normal mice were increased (Supplementary Fig. 3B). Our data confirmed that p66shc protein expression was significantly increased in the sciatic nerve of diabetic mice compared with control mice in both Western blot (Fig. 3A), and immunofluorescence data (Fig. 3B). DPN is often accompanied with by loss of sensation [33] and deficiency of p66Shc protects against cellular dysfunction caused by high glucose condition. To investigate the potential role of p66shc in DPN, von Frey test to monitor mice pain sensitivity was performed before, 4 weeks, and 8 weeks after STZ injection in p66shc knockout (KO) and wild-type (WT) mice. We proved that there is no significant difference in pain sensitivity between p66shc KO and WT mice. As expected, diabetic WT mice showed a markedly increased threshold upon exposure to von Frey filaments compared with normal WT mice, and diabetic p66shc KO mice showed a significant decrease in threshold compared with diabetic WT mice (Supplementary Fig. 3C).
Since downregulation of p66shc recovers the high glucose-induced impairment of autophagy and ER stress in SCs, p66shc may ameliorate diabetes-induced mechanical hypoalgesia
SCs are the fundamental component of peripheral nervous system as well as the neuronal regulatory network, specific in the process of peripheral nerve injury in diabetes. Excessive oxidative stress in DPN is considered a common pathway for hyperglycemia-induced SC dysfunction [34]. Oxidative stress in DPN patients is higher compared to healthy individuals and increases proportionally with disease severity. p66shc regulates oxidative stress and is implicated in diabetic diseases [35]. However, the function of p66shc in SC dysfunction is still unclear. In this study, our results revealed a vital role of p66shc in SCs in diabetes. While the impairment of autophagy was observed, p66shc was upregulated in diabetic mice’s sciatic nerve. In addition, the deletion of p66shc restored autophagy function in diabetic mice.
Sera from DPN patients induces the accumulation of autophagosomes by increasing LC3-II immune reactivity when exposed to neuroblastoma cells [36]. Autophagy was impaired in cerebellar Purkinje neurons in mice induced by STZ-induced DPN [37]. Therefore, regulation of autophagy by hyperglycemia in DPN is important for neuronal function and health. In this study, we investigated the effect of DPN-induced hyperglycemia on autophagy of SCs. In SCs, expression of p62 and LC3-II was decreased, while the presence of lysosomal inhibitor could not raise the protein level of both p62 and LC3-II. Generally, by using lysosomal inhibitors such as bafilomycin A1, the autophagic flux could be demonstrated by the raise in LC3II and p62 level in Western blot. p62 participates in the conversion of LC3-I to LC3-II and induces autophagosome maturation and autophagosome entry of damaged organelles [32]. Moreover, p62 deletion results in the impaired formation of the LC3-II, aggresome, and autophagosome, lowering cell viability under basal conditions and misfolded protein stress in cardiomyocytes [32]. LC3-I is activated by Atg7 and modified by Atg3 to bind the membrane and generate LC3-II. The LC3-II level is proportional to the number of autophagic vacuoles, and expression of LC3-II is a direct indicator of autophagy induction [38]. From our results, we suggest the downregulation of p62 caused by hyperglycemia condition was the result behind the decrease in LC3-II due to the impairment of LC3-II formation. Therefore, autophagy process was inhibited in hyperglycemia condition. Interestingly, hyperglycemia induced decrease in p62 expression and LC3-II/LC3-I ratio were reversed by p66shc knockdown in SCs.
Our data suggest a correlation with autophagy signaling pathways in the presence of oxidative stresses, including ROS. Persistent hyperglycemia during diabetes causes a redox imbalance due to overproduction of ROS. Moderate ROS levels increase autophagosome formation by converting LC3-I to LC3-II to the reduced form of Atg4
ER stress occurs when the ER processing capacity is exceeded, limits its ability to fold or assemble proteins, or after calcium depletion [42]. ER stress is activated in the peripheral nervous system of DPN mice, and chemical inducers of ER stress trigger neuralgia [43]. In this study, ER stress was increased in the sciatic nerve and high glucose-treated SCs in hyperglycemia-induced DPN mice. The increase in ER stress induced by hyperglycemia was restored by suppression of p66shc expression. This indicates that ER stress is detrimental to SC survival after DPN development, whereas inhibition of ER stress enhances SC survival. DPN-induced hyperglycemia significantly increased ER stress-related gene expression, whereas expression of basic fibroblast growth factor and nerve growth factor decreased ER stress-related gene expression. ER stress is strongly associated with autophagy [44]. To address ER stress, cells induce the unfolded protein response (UPR), which is an adaptive signaling process that triggers a variety of mechanisms to clear misfolded proteins, resolve protein folding, and enhance and maintain ER homeostasis. Autophagy under pathological ER stress, particularly neurodegeneration, is regulated by ERN1 and related pathways of the UPR. The ER degradation enhancer, mannosidase α-like 1, a target gene of XBP1 that encodes an essential component of ERAD substrate signaling and recognition, induces ER stress and thereby interferes with autophagy activation [45].
The autophagy receptor p62 prevents the effects of misfolded proteins on proteins or organelles by sequestering aggregated proteins [32,46]. Whether the UPR generates p62 bodies as a cytoprotective mechanism under ER stress is unknown. We plan to evaluate the association between decreased p62 expression and ER stress. In SCs, suppression of p66shc expression restored high glucose-induced eIF2a phosphorylation and CHOP expression but not GRP78 expression. In sciatic nerves, hyperglycemia-induced p66shc expression, but not eIF2a phosphorylation, was restored. ER stress has long been known to be linked to oxidative stress. Because protein folding is influenced by redox homeostasis, oxidative stress induces ER stress by disrupting protein folding and enhancing the production of misfolded proteins [47]. In this study, ER stress-related gene expression, which is induced by hyperglycemia, was restored by suppression of p66shc expression, albeit differently in cell culture and the mouse model.
Many stress-induced signaling pathways regulate both autophagy and apoptosis. In this study, early apoptosis of SCs was induced by high glucose, and reversed by suppression of p66shc expression. High glucose inhibits axon growth and induces apoptosis of SCs. In addition, exposure to high glucose impairs the morphology and function of SCs [48]. Oxidative stress induced by high glucose can cause DNA damage and trigger apoptosis
Overall, we demonstrate that SC dysfunction and p66shc upregulation occurred in diabetes. In addition, regulation of p66shc expression restored impaired autophagy and the functions of SCs and sciatic nerves. Therefore, agents that modulate SC autophagy may have therapeutic potential for sciatic nerve protection in diabetes.
This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2018R1A2B6007425, NRF-2018H1A2A1062309 and NRF-2014R1A6A1029617), the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare (grant number: HI21C1019) and by the research fund of Chungnam National University. Kaikobad Irani was supported by funding from R01HL147545, R01HL115955, VA Merit Award I01BX002940, and ONR N0001142012147.
None.
The authors declare no conflicts of interest.
Supplementary data including four figures can be found with this article online at https://doi.org/10.4196/kjpp.24.155
View Full Text | Article as PDF |
Abstract | Figure & Table |
Pubmed | PMC |
Print this Page | Export to Citation |
ⓒ 2019. The Korean Journal of Physiology & Pharmacology. Powered by INFOrang Co., Ltd