Letter to the Editor Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Aug 15, 2025; 16(8): 110285
Published online Aug 15, 2025. doi: 10.4239/wjd.v16.i8.110285
Microglial metabolic reprogramming: Aucubin inhibits aldose reductase to reverse diabetic neuropathic pain
Bin Li, Institution of Compared Medicine, Yangzhou University, Yangzhou 225009, Jiangsu Province, China
Bin Li, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg 66424, Saarland, Germany
ORCID number: Bin Li (0000-0003-2882-7512).
Author contributions: Li B contributed to the manuscript writing, reviewing, and editing, participated in the formal analysis, conceptualization, project administration.
Supported by the Top-level Talents Support Program of Yangzhou University; “Lv Yang Jin Feng” Outstanding Doctor of Yangzhou, No. YZLYJFJH2023YXBS169; and Natural Science Foundation of Jiangsu Province, No. BK20240907.
Conflict-of-interest statement: The authors declare that they have no conflict of interest.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Bin Li, PhD, Full Professor, Institution of Compared Medicine, Yangzhou University, No. 88 South Daxue Road, Yangzhou 225009, Jiangsu Province, China. 008480@yzu.edu.cn
Received: June 4, 2025
Revised: June 15, 2025
Accepted: July 14, 2025
Published online: August 15, 2025
Processing time: 72 Days and 7.1 Hours

Abstract

This letter critically comments on the article by Zheng et al investigating the role of aucubin in alleviating diabetic neuropathic pain (DNP). DNP arises from hyperglycaemia-induced nerve injury and microglial reprogramming toward aerobic glycolysis. Aldose reductase (also known as AKR1B1) redirects excess glucose flux through the polyol pathway, thus increasing oxidative stress and inflammation. Zheng et al show that aucubin, a plant iridoid glycoside, reverses streptozotocin-induced mechanical and thermal hypersensitivity and anxiety-like behaviour in mice. Mechanistically, aucubin restores microglial morphology, reduces glycolytic flux, enhances oxidative phosphorylation and lowers tumour necrosis factor-α, interleukin (IL)-1β and IL-6 levels in spinal tissue and cultures of the BV-2 microglial cell line. Network pharmacology and molecular docking analyses identify AKR1B1 as a key target, confirmed by the fact that short hairpin RNA knockdown of AKR1B1 eliminates the effects of aucubin. Contrary to the other studies, this study uniquely implicates the polyol pathway in microglial immunometabolism.

Key Words: Diabetic neuropathic pain; Microglia; Metabolism; Aldose reductase; Neuroinflammation

Core Tip: Zheng et al have uncovered a new anti-neuropathic function of the plant glycoside aucubin-inhibiting aldose reductase and thus preventing hyperglycaemia-driven microglial aerobic glycolysis and inflammation associated with diabetic neuropathy. This study highlights a novel metabolism-immunity axis of diabetic neuropathic pain and offers an innovative therapeutic strategy.



TO THE EDITOR

I read with great interest the recent study by Zheng et al[1], which investigates the effects of aucubin on diabetic neuropathic pain (DNP) and identifies Aldose reductase (also known as AKR1B1) as a key target.

DNP is a common, disabling complication of diabetes, affecting roughly 13%-35% of diabetic patients worldwide[2-4]. DNP arises from hyperglycemia-driven nerve injury and chronic inflammation in the peripheral and central nervous systems[5-7]. Among central mechanisms, spinal microglia have emerged as key mediators: In response to hyperglycaemia and damaged neurons, microglia undergo a metabolic shift toward glycolysis and adopt a pro-inflammatory phenotype, releasing cytokines (tumour necrosis factor-α, interleukin-1β, interleukin-6) that sensitize pain pathways[7,8]. This “immunometabolic” model of DNP is supported by recent studies showing that dysregulated microglial metabolism (e.g., increased glycolysis via hypoxia inducible factor-1α, pyruvate kinase isozyme type M2, or decreased mitochondrial function) amplifies neuroinflammation and pain[9,10].

In addition, aucubin has been shown to engage multiple intracellular signalling cascades that counteract the underlying pathogenic processes of DNP[11-15]. For example, aucubin activates the nuclear factor erythropoietin-2-related factor 2/heme oxygenase 1 antioxidant pathway, thereby suppressing reactive oxygen species (ROS) generation and the downstream nuclear factor kappa-B (NF-κB) signalling pathway in neuronal and glial cells[12]. By inhibiting the nuclear translocation of NF-κB, aucubin suppresses the transcription of pro-inflammatory genes[12]. Moreover, studies have demonstrated that aucubin modulates the members of the mitogen-activated protein kinase family (especially p38 and extracellular regulated protein kinases ½) and the phosphatidylinositol 3-kinase/protein kinase B (AKT) pathway, thus stabilizing mitochondrial function and reducing apoptotic signalling in neural tissues[11,13,15]. Finally, aucubin promotes mitophagy through the PTEN induced putative kinase 1 (PINK1)/parkin axis, thereby helping clear damaged mitochondria and restore metabolic homeostasis in microglia[14]. These convergent signalling events diminish microglial activation, decrease inflammatory cytokine release and restore oxidative phosphorylation, all of which are pivotal to alleviating DNP.

The AKR1B1 is another established contributor to diabetic complications[16,17]. Under hyperglycemia, AKR1B1 converts excess glucose to sorbitol (then fructose), causing osmotic stress, oxidative injury and pro-inflammatory signaling[18,19]. In animal models, AKR1B1 inhibitors (e.g. epalrestat) reduce sorbitol accumulation and ameliorate neuropathy, and polymorphisms in the aldose reductase gene are linked to neuropathic pain susceptibility[20-23]. Thus, AKR1B1 bridges metabolic dysregulation and inflammation in diabetes, making it a compelling target for DNP therapy.

Aucubin is a plant-derived iridoid glycoside with known anti-oxidative and anti-inflammatory properties[24,25]. Prior work has shown that aucubin alleviates inflammatory and neuropathic pain in rodents by suppressing glial activation and promoting cellular protective pathways[14,26]. For example, Yao et al[14] found that aucubin markedly reduced glial-mediated complete Freund’s adjuvant inflammatory pain via enhancing mitophagy (PINK1/parkin pathway) and lowering cytokine release. Given this background, Zheng et al’s investigation of aucubin in DNP is of high interest, as it links a natural compound, microglial metabolism and the aldose reductase pathway in the context of diabetic pain[1]. The authors report that aucubin markedly reduced pain and anxiety-like behaviors in streptozotocin (STZ)-induced diabetic mice and normalized microglial metabolism and inflammatory markers. Their integration of behavioral assays with molecular and bioinformatic analyses is commendable, and the work addresses the timely question of how metabolic regulation in microglia contributes to neuropathic pain. This letter offers a balanced discussion of the study’s strengths and limitations, focusing on mechanistic interpretation of AKR1B1’s role in microglial glycolysis and the translational implications of the findings.

STRENGTHS AND METHODOLOGICAL INNOVATIONS

Zheng et al[1] used the STZ-induced diabetes mellitus model in mice to systematically evaluate the behavioural and cellular outcomes of using aucubin in DNP treatment. They demonstrated that aucubin treatment significantly ameliorates mechanical allodynia, thermal hyperalgesia and anxiety-like behaviour in diabetic mice. These behavioural improvements are closely associated with normalization of the metabolic and inflammatory status of microglia in both STZ-treated animals and glucose-exposed microglial cultures.

Rather than detailing procedural techniques, the study effectively connects these behavioural and cellular findings to a coherent mechanistic framework. Aucubin was found to suppress microglial aerobic glycolysis and the expression of inflammatory cytokines hallmarks of neuroinflammation in DNP thereby restoring homeostasis. This supports the emerging notion that glycolytic reprogramming is a driver of microglia-mediated pain signalling in metabolic diseases.

An Innovative aspect of the study is the integration of computational and experimental strategies to identify aucubin’s molecular targets. Through network pharmacology and molecular docking, AKR1B1 was identified as the most plausible functional target, having a higher binding affinity for aucubin than matrix metalloproteinases (MMP) 2/9 do. This prediction was biologically validated by showing that aucubin downregulates AKR1B1 expression in the microglia of diabetic mice and that genetic silencing of AKR1B1 abrogates aucubin’s metabolic and anti-inflammatory effects. Thus, AKR1B1 emerges as a key node linking hyperglycaemia to microglial activation.

The study’s comprehensive design spanning in vivo phenotyping, metabolic flux assessment and gene/protein expression analyses provides robust evidence supporting the proposed mechanism. Importantly, aucubin’s effects were consistent across both in vivo and in vitro systems, strengthening its translational potential. Nevertheless, a few areas remain to be clarified. It is not yet clear whether aucubin inhibits the enzymatic activity of AKR1B1 or modulates its transcription. Additionally, the functional roles of MMP2/9 were not explored further, and the pharmacokinetics of aucubin were not evaluated, which limits insight into its clinical applicability.

In summary, the study boasts several strengths: (1) A multimodal approach linking behavior to cellular metabolism and molecular targets; (2) Identification of aldose reductase as a novel regulator of microglial glycolysis in DNP; (3) Demonstration that a natural compound with anti-oxidant properties can modify this pathway; and (4) Emphasis on both neuropathic pain and anxiety-like behaviors, reflecting the clinical spectrum of DNP. These aspects distinguish it from prior works focusing on single signaling pathways or analgesic endpoints alone.

FUTURE DIRECTIONS

We respectfully offer the following suggestions for future research to strengthen the mechanistic and translational insights.

Elucidate the aldose reductase-glycolysis pathway

Examine the metabolic consequences of AKR1B1 activity in microglia under high glucose (e.g., measure sorbitol/fructose, nicotinamide adenine dinucleotide levels) and determine how these changes signal to glycolytic enzymes or transcription factors. Investigate whether known aldose reductase replicate aucubin’s effects on microglial metabolism and inflammation.

Direct assays assessing AKR1B1 activity should be performed using purified recombinant AKR1B1 protein and one of its canonical substrates (e.g., glyceraldehyde) in the presence and absence of aucubin to determine kinetic parameters (IC50, Ki, etc.) and confirm whether aucubin acts as a competitive or non-competitive inhibitor.

Validate in primary cells and in vivo

Perform similar glycolytic flux and cytokine assays in primary microglia (mouse or human induced pluripotent stem cells-derived) and consider using conditional AKR1B1 knockout mice to assess pain and metabolic outcomes. This would confirm that findings in BV-2 cells translate to native microglia.

Assess pharmacological properties of aucubin

Determine aucubin’s blood and central nervous system bioavailability, half-life, and optimal dosing in animal models. Compare its efficacy to benchmark aldose reductase inhibitors in alleviating DNP symptoms.

Explore additional targets

Investigate the roles of MMP2, MMP9 or other predicted targets in the model, as these proteases have known involvement in pain signaling. This could clarify whether aucubin’s benefits involve multiple pathways.

Expand outcome measures

In future studies, include neuronal electrophysiology, glial markers, or imaging of neuroinflammation to see if AKR1B1 inhibition has broader effects on the nervous system, beyond microglial glycolysis.

COMPARISON WITH EXISTING LITERATURE

Zheng et al’s findings fit into a rapidly evolving landscape of pain research that emphasizes immunometabolism[1]. For example, Li et al[27] showed that Sirt3 downregulation in diabetic spinal microglia increases aerobic glycolysis and inflammation, exacerbating DNP; rescuing Sirt3 (or using metformin to block Akt/FOXO1) improved pain[26]. Similarly, Kong et al[28] identified the kinase Lyn as a promoter of glycolysis in spinal microglia after nerve injury: Lyn upregulation drove IRF5-mediated transcription of glycolytic genes and pro-inflammatory polarization, whereas a Lyn inhibitor (Bafetinib) reduced both glycolysis and pain behaviors. More recently, Hua et al[9] reported that PRMT6, a methyltransferase, is elevated in the spinal cord after nerve injury, stabilizing hypoxia inducible factor-1α and boosting pyruvate kinase isozyme type M2-driven glycolysis in microglia; PRMT6 inhibition lowered glycolysis and relieved pain and neuroinflammation[8]. Another group found that TNFAIP8 L2 (TIPE2) modulates microglial metabolism: Overexpression of TIPE2 shifted microglia from glycolysis toward oxidative phosphorylation and suppressed inflammatory activation, alleviating neuropathic pain[9].

Compared to these studies, the current work by Zheng et al[1] is novel in linking the polyol pathway to microglial immunometabolism. While the above reports highlight intrinsic cell regulators (Sirt3, Lyn, PRMT6, TIPE2) of glial metabolism, Zheng et al[1] implicates an extracellular metabolic factor: Aldose reductase. In essence, aucubin’s inhibition of AKR1B1 appears to prevent excess fructose production and ROS that would otherwise reprogram microglia toward glycolysis and inflammation. This raises a unique cross-talk: Diabetic metabolic dysregulation (polyol flux) drives central immune cell metabolism. In contrast, other interventions like curcumin broadly suppress neuroinflammation-curcumin has been shown to reduce pain hypersensitivity by inhibiting spinal microglial and astrocyte activation[29,30]-but the precise metabolic target was unclear. Aucubin’s advantage is that it addresses both systemic glucose imbalance and microglial activation. Finally, the use of a phytochemical aligns with growing interest in natural products for neuropathy; indeed, numerous reviews note that plant-derived compounds (e.g. curcumin, resveratrol, berberine) can modulate pain pathways[31]. However, few have been shown to specifically correct glial metabolism via aldose reductase. Thus this study’s novelty lies in identifying AKR1B1 as the mechanistic bridge and demonstrating a concrete effect of a plant glycoside on this axis.

CLINICAL RELEVANCE

The findings of Zheng et al[1] carry substantial translational promise for DNP. Current treatments antidepressants, antiepileptics and topical agents often provide only partial relief and bring with them significant side effects. AKR1B1 is already a validated target in diabetic complications: For example, the aldose reductase inhibitor epalrestat is used clinically in Japan to slow neuropathy progression and newer inhibitors such as ranirestat are in clinical trials. If aucubin or its optimized analogues can be shown to directly inhibit AKR1B1 (via enzyme assays) and restore microglial homeostasis in primary-cell models, while also demonstrating blood-brain barrier penetration and favourable pharmacokinetics, they can become novel therapeutic agents for DNP.

Importantly, Zheng et al[1] report that aucubin not only reverses mechanical and thermal hypersensitivity but also alleviates anxiety-like behaviour in diabetic mice, addressing the frequent comorbidity of mood disturbances in DNP. Aucubin’s established safety profile given its prevalent use in traditional medicinal herbs may accelerate its clinical development. Early-phase human trials could measure safety, target engagement (e.g., peripheral sorbitol/fructose ratios) and preliminary analgesic efficacy using quantitative sensory testing. Ultimately, combining AKR1B1 inhibition with rigorous glycaemic control could offer a dual benefit limiting both metabolic flux and neuroinflammation thereby advancing treatment beyond symptomatic relief to address core DNP pathophysiology.

CONCLUSIONS

Zheng et al[1] provide compelling evidence that aucubin mitigates DNP by suppressing AKR1B1-mediated microglial glycolysis and inflammation. We note that the authors’ original discussion of these results is comprehensive and well-contextualized, further reinforcing the validity of their conclusions. The work is scientifically rigorous and logically consistent, fitting well within the emerging literature on microglial immunometabolism. Compared to prior studies, it is novel in identifying aldose reductase as the central node linking hyperglycaemia to neuroinflammation. Clinically, it reinforces the value of targeting metabolic pathways (the polyol pathway and glycolysis) in DNP treatment. Going forward, it will be important to confirm these findings using other DNP models and to test whether aucubin directly inhibits the enzymatic activity of AKR1B1 or acts via gene regulation. In summary, this study advances our understanding of mechanisms underlying DNP and introduces a promising natural compound for use in DNP therapy.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B

Novelty: Grade A, Grade B, Grade B

Creativity or Innovation: Grade A, Grade B, Grade B

Scientific Significance: Grade A, Grade A, Grade B

P-Reviewer: Pal B; Pandurangan H S-Editor: Fan M L-Editor: A P-Editor: Xu ZH

References
1.  Zheng XZ, Yu HY, Chen YR, Fang JS. Aucubin mitigates the elevation of microglial aerobic glycolysis and inflammation in diabetic neuropathic pain via aldose reductase. World J Diabetes. 2025;16:103915.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
2.  Truini A, Spallone V, Morganti R, Tamburin S, Zanette G, Schenone A, De Michelis C, Tugnoli V, Simioni V, Manganelli F, Dubbioso R, Lauria G, Lombardi R, Jann S, De Toni Franceschini L, Tesfaye S, Fiorelli M, Spagnoli A, Cruccu G; Neuropathic Pain Special Interest Group of the Italian Society of Neurology. A cross-sectional study investigating frequency and features of definitely diagnosed diabetic painful polyneuropathy. Pain. 2018;159:2658-2666.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 39]  [Cited by in RCA: 57]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
3.  Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence and characteristics of painful diabetic neuropathy in a large community-based diabetic population in the U.K. Diabetes Care. 2011;34:2220-2224.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 505]  [Cited by in RCA: 596]  [Article Influence: 42.6]  [Reference Citation Analysis (0)]
4.  Pal B, Ghosh R, Sarkar RD, Roy GS. The irreversible, towards fatalic neuropathy: from the genesis of diabetes. Acta Diabetol. 2025;62:139-156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
5.  Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, Bril V, Russell JW, Viswanathan V. Diabetic neuropathy. Nat Rev Dis Primers. 2019;5:42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 27]  [Cited by in RCA: 143]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
6.  Sloan G, Selvarajah D, Tesfaye S. Pathogenesis, diagnosis and clinical management of diabetic sensorimotor peripheral neuropathy. Nat Rev Endocrinol. 2021;17:400-420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 84]  [Cited by in RCA: 258]  [Article Influence: 64.5]  [Reference Citation Analysis (0)]
7.  Calcutt NA. Diabetic neuropathy and neuropathic pain: a (con)fusion of pathogenic mechanisms? Pain. 2020;161:S65-S86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 89]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
8.  Milligan ED, Watkins LR. Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci. 2009;10:23-36.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1132]  [Cited by in RCA: 1070]  [Article Influence: 66.9]  [Reference Citation Analysis (0)]
9.  Hua T, Kong E, Zhang H, Lu J, Huang K, Ding R, Wang H, Li J, Han C, Yuan H. PRMT6 deficiency or inhibition alleviates neuropathic pain by decreasing glycolysis and inflammation in microglia. Brain Behav Immun. 2024;118:101-114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
10.  Li Y, Yin C, Jiang J, Yang H, Zhang F, Xing Y, Wang W, Lu C. Tumor necrosis factor α-induced protein 8-like-2 controls microglia phenotype via metabolic reprogramming in BV2 microglial cells and responses to neuropathic pain. Int J Biochem Cell Biol. 2024;169:106541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
11.  Park YJ, Lim JY, Kwon S, Shin M. Aucubin suppresses lipopolysaccharide-induced pro-inflammatory responses by blocking the NF-κB translocation signaling pathways in activated microglial cells. Phytochem Lett. 2022;48:120-127.  [PubMed]  [DOI]  [Full Text]
12.  Wang H, Zhou XM, Wu LY, Liu GJ, Xu WD, Zhang XS, Gao YY, Tao T, Zhou Y, Lu Y, Wang J, Deng CL, Zhuang Z, Hang CH, Li W. Aucubin alleviates oxidative stress and inflammation via Nrf2-mediated signaling activity in experimental traumatic brain injury. J Neuroinflammation. 2020;17:188.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 144]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
13.  Feng M, Jiang X, Zhang Q, Wang Q, She C, Li Z. Aucubin protects against retinal ganglion cell injury in diabetic rats via inhibition of the p38MAPK pathway. Am J Transl Res. 2023;15:1007-1016.  [PubMed]  [DOI]
14.  Yao D, Wang Y, Chen Y, Chen G. The Analgesia Effect of Aucubin on CFA-Induced Inflammatory Pain by Inhibiting Glial Cells Activation-Mediated Inflammatory Response via Activating Mitophagy. Pharmaceuticals (Basel). 2023;16:1545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
15.  Wang C, Cui X, Dong Z, Liu Y, Xia P, Wang X, Zhang Z, Yu S, Wu S, Liu H, Zong S, Lu Z. Attenuated memory impairment and neuroinflammation in Alzheimer's disease by aucubin via the inhibition of ERK-FOS axis. Int Immunopharmacol. 2024;126:111312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
16.  Wu TT, Chen YY, Chang HY, Kung YH, Tseng CJ, Cheng PW. AKR1B1-Induced Epithelial-Mesenchymal Transition Mediated by RAGE-Oxidative Stress in Diabetic Cataract Lens. Antioxidants (Basel). 2020;9:273.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
17.  Chen S, Li B, Chen L, Jiang H. Uncovering the mechanism of resveratrol in the treatment of diabetic kidney disease based on network pharmacology, molecular docking, and experimental validation. J Transl Med. 2023;21:380.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 49]  [Reference Citation Analysis (0)]
18.  Hotta N, Akanuma Y, Kawamori R, Matsuoka K, Oka Y, Shichiri M, Toyota T, Nakashima M, Yoshimura I, Sakamoto N, Shigeta Y. Long-term clinical effects of epalrestat, an aldose reductase inhibitor, on diabetic peripheral neuropathy: the 3-year, multicenter, comparative Aldose Reductase Inhibitor-Diabetes Complications Trial. Diabetes Care. 2006;29:1538-1544.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 249]  [Cited by in RCA: 247]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
19.  Yang Y, Zhao B, Wang Y, Lan H, Liu X, Hu Y, Cao P. Diabetic neuropathy: cutting-edge research and future directions. Signal Transduct Target Ther. 2025;10:132.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
20.  Li QR, Wang Z, Zhou W, Fan SR, Ma R, Xue L, Yang L, Li YS, Tan HL, Shao QH, Yang HY. Epalrestat protects against diabetic peripheral neuropathy by alleviating oxidative stress and inhibiting polyol pathway. Neural Regen Res. 2016;11:345-351.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 46]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
21.  Schemmel KE, Padiyara RS, D'Souza JJ. Aldose reductase inhibitors in the treatment of diabetic peripheral neuropathy: a review. J Diabetes Complications. 2010;24:354-360.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 119]  [Cited by in RCA: 135]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
22.  Hirai A, Yasuda H, Joko M, Maeda T, Kikkawa R. Evaluation of diabetic neuropathy through the quantitation of cutaneous nerves. J Neurol Sci. 2000;172:55-62.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 58]  [Cited by in RCA: 54]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
23.  Thamotharampillai K, Chan AK, Bennetts B, Craig ME, Cusumano J, Silink M, Oates PJ, Donaghue KC. Decline in neurophysiological function after 7 years in an adolescent diabetic cohort and the role of aldose reductase gene polymorphisms. Diabetes Care. 2006;29:2053-2057.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 32]  [Cited by in RCA: 23]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
24.  Jin L, Xue HY, Jin LJ, Li SY, Xu YP. Antioxidant and pancreas-protective effect of aucubin on rats with streptozotocin-induced diabetes. Eur J Pharmacol. 2008;582:162-167.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 115]  [Cited by in RCA: 126]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
25.  Jung E, Park SB, Jung WK, Kim HR, Kim J. Aucubin, An Active Ingredient in Aucuba japonica, Prevents N-methyl-N-nitrosourea-induced Retinal Degeneration in Mice. Molecules. 2019;24:4437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
26.  Yang P, Zhang Q, Shen H, Bai X, Liu P, Zhang T. Research progress on the protective effects of aucubin in neurological diseases. Pharm Biol. 2022;60:1088-1094.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
27.  Li Y, Kong E, Ding R, Chu R, Lu J, Deng M, Hua T, Yang M, Wang H, Chen D, Song H, Wei H, Zhang P, Han C, Yuan H. Hyperglycemia-induced Sirt3 downregulation increases microglial aerobic glycolysis and inflammation in diabetic neuropathic pain pathogenesis. CNS Neurosci Ther. 2024;30:e14913.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
28.  Kong E, Li Y, Ma P, Zhang Y, Ding R, Hua T, Yang M, Yuan H. Lyn-mediated glycolysis enhancement of microglia contributes to neuropathic pain through facilitating IRF5 nuclear translocation in spinal dorsal horn. J Cell Mol Med. 2023;27:1664-1681.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
29.  Xie W, Xie W, Kang Z, Jiang C, Liu N. Administration of Curcumin Alleviates Neuropathic Pain in a Rat Model of Brachial Plexus Avulsion. Pharmacology. 2019;103:324-332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
30.  Liu S, Li Q, Zhang MT, Mao-Ying QL, Hu LY, Wu GC, Mi WL, Wang YQ. Curcumin ameliorates neuropathic pain by down-regulating spinal IL-1β via suppressing astroglial NALP1 inflammasome and JAK2-STAT3 signalling. Sci Rep. 2016;6:28956.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 52]  [Cited by in RCA: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
31.  Hasriadi, Dasuni Wasana PW, Vajragupta O, Rojsitthisak P, Towiwat P. Mechanistic Insight into the Effects of Curcumin on Neuroinflammation-Driven Chronic Pain. Pharmaceuticals (Basel). 2021;14:777.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]