Abstract
While there is ample evidence indicating an increased occurrence of general neurological conditions among individuals with diabetes, there has been limited exploration into the cause-and-effect connection between type 2 diabetes (T2D) and specific neurological disorders, including conditions like carpal tunnel syndrome and Bell’s palsy. We used Mendelian randomization (MR) approach to investigate the causal effects of T2D on 67 neurological diseases. We primarily utilized the inverse-variance weighted method for the analysis, and also employed the weighted median and MR-Egger methods in our study. To detect and correct potential outliers, MR-PRESSO analysis was used. Heterogeneity was assessed using Cochrane Q-values. The MR analyses found a possible relationship between T2D and a risk increase of 8 diseases at suggestive level of evidence (P < .05). Notably, among the positive findings that met the false discovery rate threshold, nerve, nerve root, and plexus disorders (odds ratio [OR] = 1.11; 95% confidence interval [CI] = 1.08–1.15); neurological diseases (OR = 1.05; 95% CI = 1.03–1.07) and carpal tunnel syndrome (OR = 1.10; 95% CI = 1.05–1.16) were identified. Our findings affirm a cause-and-effect association between T2D and certain neurological disorders.
Keywords: carpal tunnel syndrome, causal relationships, Mendelian randomization, neurological disorders, type 2 diabetes
1. Introduction
Neurological issues are one of the leading causes of death globally, following cardiovascular ailments.[1–4] Over the 30 years preceding up to 2020, there was a 39% increase in mortality from neurological disorders and a 15% increase in disability-adjusted life years.[2,5] The most prevalent contributors to disability-adjusted life years include stroke, migraine, Alzheimer disease, and meningitis.[1,2,6–9] Likewise, diabetes affects around 10% of individuals globally, with type 2 diabetes (T2D) accounting for 90% of cases.[10–12] Diabetes has numerous risk factors[13–16] and is associated with a variety of complications.[17–22] The brain is extremely glucose sensitive, with its levels influencing neuronal maintenance, neurogenesis, neurotransmitter modulation, and plasticity of synapses.[23,24] It is anticipated that between 30% and 50% of diabetics may acquire neuropathy.[25,26] Other studies also have discovered a relationship between neurological disorders and problems with blood glucose metabolism. For instance, based on other Mendelian randomization (MR) studies, T2D is independently associated with carpal tunnel syndrome (CTS).[27] A case–control study, on the other hand, did not produce compelling evidence of an independent connection between T2D and CTS.[28] Observational studies are susceptible to confounding variables and reverse causation, and the effect of T2D on specific type of neurologic illnesses remains an inconsistency.
MR using genetic variation as an instrumental variable (IVs) helps to clarify these associations.[29,30] It efficiently reduces potential confounding and reverse causality interference by following the random allocation of alleles during conception and the absence of genotype modification by illnesses.[31,32] With the causal mechanisms of T2D in causing neurological disorders still unknown, our study used a two-sample MR approach to assess the probability of neurological diseases in T2D patients. Our study may contribute to discovering novel strategies to prevent neurological complications in individuals with T2D.
2. Methods
With MR analysis, we assessed the causal relationship between T2D and 67 neurological disorders, utilizing information gathered through genome-wide association study (GWAS) of single nucleotide polymorphisms (SNPs) that were significantly related to T2D. The GWAS summary statistics pertaining to T2D was extracted from the study conducted by Anubha Mahajan et al,[33] while data for the dependent variables was acquired from all 67 neurological diseases (further details are provided in Table S1, Supplemental Digital Content, http://links.lww.com/MD/N866 accessible in the public database).[34,35]
We applied several criteria to select the IVs for our MR analyses, as described previously[36,37]: (1) IVs are tightly connected with exposure on a genetic basis (P < 5 × 10-8), (2) to eliminate linkage disequilibrium in IVs, we used R2 < 0.001, a distance of 10 Mb, (3) along with a minor allele frequency criterion of 0.01. F-statistics were also employed to evaluate the IVs’ strength: values >10 were considered indicative of a lower probability of weak IVs.[38]
The inverse-variance weighted method was the principal method used in this study, and we also employed the weighted median[39] and MR-Egger[40] methods in this study, similar to our previous works.[41–43] To detect and eliminate any outliers, we employed MR-PRESSO analysis.[44] Heterogeneity was evaluated through the examination of Cochrane Q-values. Moreover, using the leave-one-out method, we iteratively excluded individual SNPs to assess whether the results remain significant.[45] The strength of causal effect was evaluated by analyzing odds ratios (OR) and 95% confidence intervals (95% CI). To account for multiple testing, a false discovery rate (FDR) threshold of 5% was applied. The significance threshold adjusted for FDR is presented in Figure 1. Indications of suggestive associations were taken into account when the P-value was <.05. The MR analyses were conducted with the two-sample MR software package in R (TwoSampleMR version 0.6.4).[46,47]
Figure 1.
The distribution of P-values for the associations between T2D and 67 neurologic disorders in the MR analysis. The threshold line 1 showed the significance threshold adjusted for FDR. The threshold line 2 showed the significance threshold of P = .05. FDR = false discovery rate, MR = Mendelian randomization, T2D = type 2 diabetes.
3. Results
3.1. Results of the MR analysis
A total of 184 SNPs were chosen as IVs for T2D, with all these IVs having F-statistics topping 10 (refer Table S2, Supplemental Digital Content, http://links.lww.com/MD/N866). The inverse-variance weighted approach results suggested a possible causal relationship between T2D and 8 neurological disorders, such as “Carpal tunnel syndrome” (OR = 1.10; 95% CI: 1.05–1.16), “Bell’s palsy” (OR = 1.15; 95% CI: 1.05–1.27), “Lesion of sciatic nerve” (OR = 1.18; 95% CI: 1.04–1.35), “Transient ischemic attack” (OR = 1.05; 95% CI: 1.00–1.09), and “Cervical root disorders” (OR = 1.39; 95% CI: 1.00–1.94) (Figs. 1 and 2; Table S3, Supplemental Digital Content, http://links.lww.com/MD/N866). Furthermore, substantial associations between T2D and ailments such as “Nerve, nerve root, and plexus disorders,” “Neurological diseases,” and “Carpal tunnel syndrome” remained significant even after the FDR correction. Using the MR-Egger and weighted median techniques, the relationships between T2D and 8 out of 67 neurological disorders showed similar directions (see Fig. 2 and Table S3, Supplemental Digital Content, http://links.lww.com/MD/N866). Figure 3 shows a scatter plot illustrating the causative relationships between T2D and 8 neurological illnesses.
Figure 2.
Causal effects of T2D on 8 neurologic disorders in the MR analysis, showing odds ratios and corresponding 95% confidence intervals. IVW = inverse-variance weighted, MR = Mendelian randomization, T2D = type 2 diabetes, WM = weighted median.
Figure 3.
Scatter plot indicating the causal associations between T2D and 8 neurologic disorders. MR = Mendelian randomization, SNP = single nucleotide polymorphism, T2D = type 2 diabetes.
3.2. Results of the sensitivity analysis
The potential heterogeneity was examined (Fig. 4; Table S4, Supplemental Digital Content, http://links.lww.com/MD/N866), and the findings from Figure S1, Supplemental Digital Content, http://links.lww.com/MD/N866 suggest the removal of the majority of SNPs in the leave-one-out analysis had no significant effect on the outcomes. As shown in Table S5, Supplemental Digital Content, http://links.lww.com/MD/N866, the MR-Egger approach revealed no evidence of horizontal pleiotropy. Despite the MR-PRESSO study found several outliers, adjustments made little difference to the results (Table S6, Supplemental Digital Content, http://links.lww.com/MD/N866).
Figure 4.
Funnel plot of the analyses investigating the causal effects of T2D on 8 neurologic disorders, with each SNP functioning as an IV. IV = instrumental variable, MR = Mendelian randomization, SE = standard error, SNP = single nucleotide polymorphism, T2D = type 2 diabetes.
4. Discussion
We investigated the causative impact of T2D on 67 different neurologic disorders thorough MR analyses. Our data reveal that T2D is suggestively connected with an increased risk of 8 illnesses, such as “Bell’s palsy,” “Lesion of sciatic nerve” and “Cervical root disorders.” Despite FDR correction, genetic predisposition to T2D remained strongly related with an elevated risk of “nerve, nerve root, and plexus disorders,” “neurological diseases,” and “carpal tunnel syndrome.”
T2D was found to be strongly associated with a range of brain problems in a longitudinal investigation, including several psychiatric ailments and some neurological diseases (inflammatory brain diseases, epilepsy, and migraines).[48] A cross-sectional study of 366 persons with T2D indicated that the prevalence of peripheral neuropathy was as high as 53.6%.[49] Cervical root abnormalities, Bell’s palsy, sciatic nerve lesion, and CTS were found as types of peripheral neuropathy in our investigation. Furthermore, 39.3% (139 people) of 353 T2D patients had CTS within a year.[50] The findings of our investigation are consistent with the results of previous studies.
Neurons require glucose,[51,52] and in diabetics, this requirement is increased fourfold.[53,54] Because glucose uptake in neurons is independent of insulin, high glucose levels saturate hexokinase and shift more than 30% of glucose to the polyol pathway, resulting in increased NADPH consumption.[55–58] It damages proteins via irreversible glycation, reduces glutathione peroxidase’s capacity for metabolism, and causes oxidative stress in neurons.[56,59] Oxidative stress can lead to cell apoptosis,[60,61] and studies demonstrate the involvement of oxidative stress in neurological conditions.[62–66] Oxidative stress also activates mitogen-activated protein kinases like p38, c-Jun NH2-terminal kinase, and extracellular signal-regulated kinase, and the consequent over-phosphorylation of cells can induce neurodegenerative diseases.[67–69]
High glucose-induced protein glycation has a significant influence on the nervous system.[70,71] T2D causes a glycation process, which results in the production of advanced glycation end products (AGEs).[72–74] The receptor for advanced glycation end products (RAGE)-mediated transmission system causes neuronal stress and activates several signaling pathways, affecting nerve cell activity.[75,76] RAGE could interact and bind with AGEs.[77–79] RAGE binding to AGEs increases endothelial permeability to macromolecules and stimulates the synthesis of vascular cell adhesion molecule-1, tissue factor, and interleukin-6,[80–82] which play important role in inflammation.[83–86] As consequence, detrimental impacts on protein structure and function ensue. Glycation is involved in amyotrophic lateral sclerosis, Parkinson disease, and Alzheimer disease.[87–89]
The fibrous canal through which the median nerve passes can cause CTS, a compressive peripheral neuropathy resulting from nerve compression.[90,91] Increased glucose levels make peripheral nerves more vulnerable to compression.[71,92] Diabetes has been linked to decreased myelinated nerve fiber density in the posterior interosseous nerve, as well as thickening of the basal lamina of nerve endoneurial capillaries and decreased capillary density, making diabetic patients more susceptible to microvascular compression.[93,94] Patients with CTS and diabetes who experience pathological ischemia–reperfusion (I/R) also demonstrate microvascular ischemia in nerve vasculature.[50,95] Moreover, diabetes stimulates the production of profibrotic factors associated with sub-synovial connective tissue, such as vascular endothelial growth factor and interleukins, resulting in an increase in fibrosis of the sub-synovial connective tissue, which is a characteristic of CTS initiation and progression.[96,97]
Regarding our other findings, there is currently a dearth of research on a link between T2D and cervical root disorders. Furthermore, additional observational or MR studies have shown that T2D is not associated with a rise in the risk of Bell’s palsy.[98,99] However, our MR data suggest a probable causal connection between T2D and an increased frequency of cervical root issues and Bell’s palsy. Hence, we believe it is worthy of further investigation in the future.
According to our viewpoint, this is the first systematic MR study explaining the causal relationship between T2D and a variety of neurological diseases, providing more solid evidence of causation than observational studies. The impact of confounding factors may be considerably decreased by random allocation, and the genome stays unchanged by the disease, thereby reducing causal disruption. Moreover, the widespread availability of publicly known genetic associations enables the selection of appropriate genetic IVs, making MR an effective and affordable technique.[100]
Our research, however, has certain limitations. Firstly, associations between exposures and outcomes may differ among populations. Since our findings are primarily derived from a European population, their applicability to other groups may be limited. Secondly, because we used GWAS summary data, we could not stratify the data by variables like age, which may have introduced population bias.
5. Conclusion
T2D has been identified as a risk factor for several types of neurological conditions, such as CTS. Understanding how T2D affects the nervous system may lead to the discovery of novel therapeutic targets, which reduces the risk of neurological illness development and progression.
Author contributions
Data curation: Yongfang Wei.
Formal analysis: Zhaoquan Wu.
Methodology: Mengling Zhang.
Writing – original draft: Yongfang Wei, Shuling Xu, Zhaoquan Wu, Mengling Zhang, Meihua Bao, Binsheng He.
Writing – review & editing: Meihua Bao, Binsheng He.
Supplementary Material
Abbreviations:
- AGEs
- advanced glycation end products
- CIs
- confidence intervals
- CTS
- carpal tunnel syndrome
- FDR
- false discovery rate
- GWAS
- genome-wide association study
- I/R
- ischemia–reperfusion
- IVs
- instrumental variables
- MR
- Mendelian randomization
- ORs
- odds ratios
- RAGEs
- receptor for advanced glycation end products
- SNPs
- single nucleotide polymorphism
- T2D
- type 2 diabetes
This study was supported by the “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science and Clinical Medicine), and the Key Research Projects of Hunan Provincial Department of Education (23A0665).
The GWASs included in this work were approved by their relevant review board, and informed consent were given by all participants.
The authors have no conflicts of interest to disclose.
The datasets generated during and/or analyzed during the current study are publicly available.
Supplemental Digital Content is available for this article.
How to cite this article: Wei Y, Xu S, Wu Z, Zhang M, Bao M, He B. Exploring the causal relationships between type 2 diabetes and neurological disorders using a Mendelian randomization strategy. Medicine 2024;103:46(e40412).
YW and SX contributed equally to this work.
Contributor Information
Yongfang Wei, Email: yongfangweicsmu@163.com.
Zhaoquan Wu, Email: wuzq900815@163.com.
Mengling Zhang, Email: zml17267897219@163.com.
Meihua Bao, Email: mhbao78@163.com.
Binsheng He, Email: hnaios@163.com.
References
- [1].Deuschl G, Beghi E, Fazekas F, et al. The burden of neurological diseases in Europe: an analysis for the global burden of disease study 2017. Lancet Public Health. 2020;5:e551–67. [DOI] [PubMed] [Google Scholar]
- [2].Feigin VL, Vos T, Nichols E, et al. The global burden of neurological disorders: translating evidence into policy. Lancet Neurol. 2020;19:255–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Liang D, Guan Q, Huang M, et al. Changing trends of disease burden of stroke from 1990 to 2019 and its predictions among the Chinese population. Front Neurol. 2023;14:1255524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Lou Z, Wang AP, Duan XM, Hu GH, Zuo ML, Yang ZB. Role of ALK5/SMAD2/3 signaling in the regulation of NOX expression in cerebral ischemia/reperfusion injury. Exp Ther Med. 2018;16:1671–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].GBD 2016 Neurology Collaborators. Global, regional, and national burden of neurological disorders, 1990-2016: a systematic analysis for the global burden of disease study 2016. Lancet Neurol. 2019;18:459–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Wang Y, Liang J, Fang Y, et al. Burden of common neurologic diseases in Asian Countries, 1990-2019: an analysis for the global burden of disease study 2019. Neurology. 2023;100:e2141–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Liu Y, Li H, Wang X, et al. Anti-Alzheimers molecular mechanism of icariin: insights from gut microbiota, metabolomics, and network pharmacology. J Transl Med. 2023;21:277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Fan Z, He Y, Sun W, Li Z, Ye C, Wang C. Amoxicillin-induced aseptic meningitis: clinical features, diagnosis and management. Eur J Med Res. 2023;28:301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Kong F, Li L, Zhang D, et al. Healthy adults with Streptococcus pneumoniae meningitis and Streptococcus pneumoniae subdural abscess: two case reports and a literature review. J Int Med Res. 2022;50:3000605221137470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018;14:88–98. [DOI] [PubMed] [Google Scholar]
- [11].Chatterjee S, Khunti K, Davies MJ. Type 2 diabetes. Lancet. 2017;389:2239–51. [DOI] [PubMed] [Google Scholar]
- [12].Liang D, Cai X, Guan Q, Ou Y, Zheng X, Lin X. Burden of type 1 and type 2 diabetes and high fasting plasma glucose in Europe, 1990-2019: a comprehensive analysis from the global burden of disease study 2019. Front Endocrinol (Lausanne). 2023;14:1307432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Zhou Y, Chai X, Yang G, Sun X, Xing Z. Changes in body mass index and waist circumference and heart failure in type 2 diabetes mellitus. Front Endocrinol (Lausanne). 2023;14:1305839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Schmittdiel J, Vijan S, Fireman B, Lafata JE, Oestreicher N, Selby JV. Predicted quality-adjusted life years as a composite measure of the clinical value of diabetes risk factor control. Med Care. 2007;45:315–21. [DOI] [PubMed] [Google Scholar]
- [15].Leong A, Rahme E, Dasgupta K. Spousal diabetes as a diabetes risk factor: a systematic review and meta-analysis. BMC Med. 2014;12:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Li JM, Li X, Chan LWC, et al. Lipotoxicity-polarised macrophage-derived exosomes regulate mitochondrial fitness through Miro1-mediated mitophagy inhibition and contribute to type 2 diabetes development in mice. Diabetologia. 2023;66:2368–86. [DOI] [PubMed] [Google Scholar]
- [17].Yang YY, Shi LX, Li JH, Yao LY, Xiang DX. Piperazine ferulate ameliorates the development of diabetic nephropathy by regulating endothelial nitric oxide synthase. Mol Med Rep. 2019;19:2245–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Xu Z, Zhang P, Chen Y, Jiang J, Zhou Z, Zhu H. Comparing SARC-CalF with SARC-F for screening sarcopenia in adults with type 2 diabetes mellitus. Front Nutr. 2022;9:803924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Luo M, Cao Q, Wang D, et al. The impact of diabetes on postoperative outcomes following spine surgery: a meta-analysis of 40 cohort studies with 2.9 million participants. Int J Surg. 2022;104:106789. [DOI] [PubMed] [Google Scholar]
- [20].Yu T, Xu B, Bao M, et al. Identification of potential biomarkers and pathways associated with carotid atherosclerotic plaques in type 2 diabetes mellitus: a transcriptomics study. Front Endocrinol (Lausanne). 2022;13:981100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Su M, Hu R, Tang T, Tang W, Huang C. Review of the correlation between Chinese medicine and intestinal microbiota on the efficacy of diabetes mellitus. Front Endocrinol (Lausanne). 2022;13:1085092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Chen J, Li X, Liu H, et al. Bone marrow stromal cell-derived exosomal circular RNA improves diabetic foot ulcer wound healing by activating the nuclear factor erythroid 2-related factor 2 pathway and inhibiting ferroptosis. Diabet Med. 2023;40:e15031. [DOI] [PubMed] [Google Scholar]
- [23].Rahman MH, Peng S, Hu X, et al. A network-based bioinformatics approach to identify molecular biomarkers for type 2 diabetes that are linked to the progression of neurological diseases. Int J Environ Res Public Health. 2020;17:1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Dienel GA. Brain glucose metabolism: integration of energetics with function. Physiol Rev. 2019;99:949–1045. [DOI] [PubMed] [Google Scholar]
- [25].Zimmerman M, Gottsäter A, Dahlin LB. Carpal tunnel syndrome and diabetes-a comprehensive review. J Clin Med. 2022;11:1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Tesfaye S, Selvarajah D. Advances in the epidemiology, pathogenesis and management of diabetic peripheral neuropathy. Diabetes Metab Res Rev. 2012;28(S1):8–14. [DOI] [PubMed] [Google Scholar]
- [27].Mi J, Liu Z. Obesity, type 2 diabetes, and the risk of carpal tunnel syndrome: a two-sample mendelian randomization study. Front Genet. 2021;12:688849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Hendriks SH, van Dijk PR, Groenier KH, Houpt P, Bilo HJ, Kleefstra N. Type 2 diabetes seems not to be a risk factor for the carpal tunnel syndrome: a case control study. BMC Musculoskelet Disord. 2014;15:346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Sanderson E, Glymour MM, Holmes MV, et al. Mendelian randomization. Nat Rev Methods Primers. 2022;2:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].von Hinke Kessler Scholder S, Smith GD, Lawlor DA, Propper C, Windmeijer F. Mendelian randomization: the use of genes in instrumental variable analyses. Health Econ. 2011;20:893–6. [DOI] [PubMed] [Google Scholar]
- [31].Davey Smith G, Hemani G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum Mol Genet. 2014;23:R89–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Carter AR, Sanderson E, Hammerton G, et al. Mendelian randomisation for mediation analysis: current methods and challenges for implementation. Eur J Epidemiol. 2021;36:465–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Mahajan A, Taliun D, Thurner M, et al. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat Genet. 2018;50:1505–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature. 2023;613:508–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Elsworth B, Lyon M, Alexander T, et al. The MRC IEU OpenGWAS data infrastructure. bioRxiv. 2020:2020.08.10.244293. [Google Scholar]
- [36].Chen R, Xu S, Ding Y, et al. Dissecting causal associations of type 2 diabetes with 111 types of ocular conditions: a Mendelian randomization study. Front Endocrinol (Lausanne). 2023;14:1307468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Fu Q, Chen R, Xu S, et al. Assessment of potential risk factors associated with gestational diabetes mellitus: evidence from a Mendelian randomization study. Front Endocrinol (Lausanne). 2023;14:1276836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Burgess S, Thompson SG. Avoiding bias from weak instruments in Mendelian randomization studies. Int J Epidemiol. 2011;40:755–64. [DOI] [PubMed] [Google Scholar]
- [39].Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40:304–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Burgess S, Thompson SG. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32:377–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Fu Q, Chen R, Ding Y, et al. Sodium intake and the risk of various types of cardiovascular diseases: a Mendelian randomization study. Front Nutr. 2023;10:1250509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Jiang Y, Chen R, Xu S, et al. Assessing causal associations of hyperparathyroidism with blood counts and biochemical indicators: a Mendelian randomization study. Front Endocrinol (Lausanne). 2023;14:1295040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Jiang Y, Chen R, Xu S, et al. Endocrine and metabolic factors and the risk of idiopathic pulmonary fibrosis: a Mendelian randomization study. Front Endocrinol (Lausanne). 2023;14:1321576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50:693–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Burgess S, Davey Smith G, Davies NM, et al. Guidelines for performing Mendelian randomization investigations: update for summer 2023. Wellcome Open Res. 2019;4:186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Hemani G, Zheng J, Elsworth B, et al. The MR-Base platform supports systematic causal inference across the human phenome. Elife. 2018;7:e34408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Hemani G, Tilling K, Davey Smith G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 2017;13:e1007081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Wimberley T, Horsdal HT, Brikell I, et al. Temporally ordered associations between type 2 diabetes and brain disorders - a Danish register-based cohort study. BMC Psychiatry. 2022;22:573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Abdissa D, Hamba N, Kene K, et al. Prevalence and determinants of peripheral neuropathy among type 2 adult diabetes patients attending Jimma University Medical Center, Southwest Ethiopia, 2019, an institutional-based cross-sectional study. J Diabetes Res. 2020;2020:9562920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Dong D, Liu H. Prevalence of carpal tunnel syndrome in patients with long-term type 2 diabetes mellitus. Heliyon. 2022;8:e12615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Li H, Guglielmetti C, Sei YJ, et al. Neurons require glucose uptake and glycolysis in vivo. Cell Rep. 2023;42:112335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Mergenthaler P, Lindauer U, Dienel GA, Meisel A. Sugar for the brain: the role of glucose in physiological and pathological brain function. Trends Neurosci. 2013;36:587–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Tomlinson DR, Gardiner NJ. Glucose neurotoxicity. Nat Rev Neurosci. 2008;9:36–45. [DOI] [PubMed] [Google Scholar]
- [54].Wang R, Liu X, Hentges ST, et al. The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes. 2004;53:1959–65. [DOI] [PubMed] [Google Scholar]
- [55].Emmanuel Y, Cochlin LE, Tyler DJ, de Jager CA, Smith AD, Clarke K. Human hippocampal energy metabolism is impaired during cognitive activity in a lipid infusion model of insulin resistance. Brain Behav. 2013;3:134–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Chung SS, Ho EC, Lam KS, Chung SK. Contribution of polyol pathway to diabetes-induced oxidative stress. J Am Soc Nephrol. 2003;14(8 Suppl 3):S233–6. [DOI] [PubMed] [Google Scholar]
- [57].Yagihashi S, Yamagishi SI, Wada Ri R, et al. Neuropathy in diabetic mice overexpressing human aldose reductase and effects of aldose reductase inhibitor. Brain. 2001;124(Pt 12):2448–58. [DOI] [PubMed] [Google Scholar]
- [58].Holoman NC, Aiello JJ, Trobenter TD, et al. Reduction of Glut1 in the neural retina but not the RPE alleviates polyol accumulation and normalizes early characteristics of diabetic retinopathy. J Neurosci. 2021;41:3275–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Tigchelaar C, van Zuylen ML, Hulst AH, et al. Elevated cerebrospinal fluid glucose levels and diabetes mellitus are associated with activation of the neurotoxic polyol pathway. Diabetologia. 2022;65:1098–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Chen Y, Tan S, Liu M, Li J. LncRNA TINCR is downregulated in diabetic cardiomyopathy and relates to cardiomyocyte apoptosis. Scand Cardiovasc J. 2018;52:335–9. [DOI] [PubMed] [Google Scholar]
- [61].Kannan K, Jain SK. Oxidative stress and apoptosis. Pathophysiology. 2000;7:153–63. [DOI] [PubMed] [Google Scholar]
- [62].Hassan W, Noreen H, Rehman S, Kamal MA, da Rocha JBT. Association of oxidative stress with neurological disorders. Curr Neuropharmacol. 2022;20:1046–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Tang L, Wang Y, Xiang J, et al. lncRNA and circRNA expression profiles in the hippocampus of Aβ(25-35)-induced AD mice treated with Tripterygium glycoside. Exp Ther Med. 2023;26:426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative stress and antioxidants in neurodegenerative disorders. Antioxidants (Basel). 2023;12:517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Houldsworth A. Role of oxidative stress in neurodegenerative disorders: a review of reactive oxygen species and prevention by antioxidants. Brain Commun. 2024;6:fcad356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Huo R, Liu Y, Xu H, et al. Associations between carotid atherosclerotic plaque characteristics determined by magnetic resonance imaging and improvement of cognition in patients undergoing carotid endarterectomy. Quant Imaging Med Surg. 2022;12:2891–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Purves T, Middlemas A, Agthong S, et al. A role for mitogen-activated protein kinases in the etiology of diabetic neuropathy. FASEB J. 2001;15:2508–14. [DOI] [PubMed] [Google Scholar]
- [68].Chen L, Liu L, Yin J, Luo Y, Huang S. Hydrogen peroxide-induced neuronal apoptosis is associated with inhibition of protein phosphatase 2A and 5, leading to activation of MAPK pathway. Int J Biochem Cell Biol. 2009;41:1284–95. [DOI] [PubMed] [Google Scholar]
- [69].Fan Z, Wang X, Zhang M, Zhao C, Mei C, Li P. MAPK pathway inhibitors attenuated hydrogen peroxide induced damage in neural cells. Biomed Res Int. 2019;2019:5962014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].King RH. The role of glycation in the pathogenesis of diabetic polyneuropathy. Mol Pathol. 2001;54:400–8. [PMC free article] [PubMed] [Google Scholar]
- [71].Rom S, Heldt NA, Gajghate S, Seliga A, Reichenbach NL, Persidsky Y. Hyperglycemia and advanced glycation end products disrupt BBB and promote occludin and claudin-5 protein secretion on extracellular microvesicles. Sci Rep. 2020;10:7274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Indyk D, Bronowicka-Szydełko A, Gamian A, Kuzan A. Advanced glycation end products and their receptors in serum of patients with type 2 diabetes. Sci Rep. 2021;11:13264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Goh S-Y, Cooper ME. The role of advanced glycation end products in progression and complications of diabetes. J Clin Endocrinol Metab. 2008;93:1143–52. [DOI] [PubMed] [Google Scholar]
- [74].Sergi D, Boulestin H, Campbell FM, Williams LM. The role of dietary advanced glycation end products in metabolic dysfunction. Mol Nutr Food Res. 2021;65:1900934. [DOI] [PubMed] [Google Scholar]
- [75].Chegão A, Vicente Miranda H. Unveiling new secrets in Parkinson’s disease: the glycatome. Behav Brain Res. 2023;442:114309. [DOI] [PubMed] [Google Scholar]
- [76].Bongarzone S, Savickas V, Luzi F, Gee AD. Targeting the Receptor for Advanced Glycation Endproducts (RAGE): a medicinal chemistry perspective. J Med Chem. 2017;60:7213–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Khalid M, Petroianu G, Adem A. Advanced glycation end products and diabetes mellitus: mechanisms and perspectives. Biomolecules. 2022;12:542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Ramasamy R, Yan SF, Schmidt AM. Receptor for AGE (RAGE): signaling mechanisms in the pathogenesis of diabetes and its complications. Ann N Y Acad Sci. 2011;1243:88–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Teissier T, Boulanger E. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology. 2019;20:279–301. [DOI] [PubMed] [Google Scholar]
- [80].Schmidt AM, Hasu M, Popov D, et al. Receptor for advanced glycation end products (AGEs) has a central role in vessel wall interactions and gene activation in response to circulating AGE proteins. Proc Natl Acad Sci U S A. 1994;91:8807–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Schmidt AM, Hori O, Chen JX, et al. Advanced glycation endproducts interacting with their endothelial receptor induce expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured human endothelial cells and in mice. A potential mechanism for the accelerated vasculopathy of diabetes. J Clin Invest. 1995;96:1395–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Wautier JL, Zoukourian C, Chappey O, et al. Receptor-mediated endothelial cell dysfunction in diabetic vasculopathy. Soluble receptor for advanced glycation end products blocks hyperpermeability in diabetic rats. J Clin Invest. 1996;97:238–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Kong DH, Kim YK, Kim MR, Jang JH, Lee S. Emerging roles of vascular cell adhesion molecule-1 (VCAM-1) in immunological disorders and cancer. Int J Mol Sci. 2018;19:1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014;6:a016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Zhang Y, Meng Y, Chen M, et al. Correlation between the systemic immune-inflammation indicator (SII) and serum ferritin in US adults: a cross-sectional study based on NHANES 2015-2018. Ann Med. 2023;55:2275148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Cui Y, Wang X, Lin F, et al. MiR-29a-3p improves acute lung injury by reducing alveolar epithelial cell PANoptosis. Aging Dis. 2022;13:899–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Vicente Miranda H, Outeiro TF. The sour side of neurodegenerative disorders: the effects of protein glycation. J Pathol. 2010;221:13–25. [DOI] [PubMed] [Google Scholar]
- [88].Vicente Miranda H, El-Agnaf OMA, Outeiro TF. Glycation in Parkinson’s disease and Alzheimer’s disease. Mov Disord. 2016;31:782–90. [DOI] [PubMed] [Google Scholar]
- [89].Ray R, Juranek JK, Rai V. RAGE axis in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis. Neurosci Biobehav Rev. 2016;62:48–55. [DOI] [PubMed] [Google Scholar]
- [90].Padua L, Coraci D, Erra C, et al. Carpal tunnel syndrome: clinical features, diagnosis, and management. Lancet Neurol. 2016;15:1273–84. [DOI] [PubMed] [Google Scholar]
- [91].Raizman NM, Blazar PE. AAOS appropriate use criteria: management of carpal tunnel syndrome. J Am Acad Orthop Surg. 2018;26:e131–3. [DOI] [PubMed] [Google Scholar]
- [92].Rydberg M, Zimmerman M, Gottsäter A, Nilsson PM, Melander O, Dahlin LB. Diabetes mellitus as a risk factor for compression neuropathy: a longitudinal cohort study from southern Sweden. BMJ Open Diabetes Res Care. 2020;8:e001298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Olsson C, Rydberg M, Zimmerman M. Diabetic retinopathy as a predictor for peripheral compression neuropathies, a registry-based study. PLoS One. 2022;17:e0275598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Thomsen NO, Mojaddidi M, Malik RA, Dahlin LB. Reduced myelinated nerve fibre and endoneurial capillary densities in the forearm of diabetic and non-diabetic patients with carpal tunnel syndrome. Acta Neuropathol. 2009;118:785–91. [DOI] [PubMed] [Google Scholar]
- [95].Schratzberger P, Walter DH, Rittig K, et al. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J Clin Invest. 2001;107:1083–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Sharma D, Jaggi AS, Bali A. Clinical evidence and mechanisms of growth factors in idiopathic and diabetes-induced carpal tunnel syndrome. Eur J Pharmacol. 2018;837:156–63. [DOI] [PubMed] [Google Scholar]
- [97].Samii A, Unger J, Lange W. Vascular endothelial growth factor expression in peripheral nerves and dorsal root ganglia in diabetic neuropathy in rats. Neurosci Lett. 1999;262:159–62. [DOI] [PubMed] [Google Scholar]
- [98].Liu H, Sun Q, Bi W, Mu X, Li Y, Hu M. Genetic association of hypertension and several other metabolic disorders with Bell’s palsy. Front Genet. 2023;14:1077438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Şevik Eliçora S, Erdem D. Does type 2 diabetes mellitus affect the healing of Bell’s Palsy in adults? Can J Diabetes. 2018;42:433–6. [DOI] [PubMed] [Google Scholar]
- [100].Sekula P, Del Greco MF, Pattaro C, Köttgen A. Mendelian randomization as an approach to assess causality using observational data. J Am Soc Nephrol. 2016;27:3253–65. [DOI] [PMC free article] [PubMed] [Google Scholar]