Next Article in Journal
RIPK1-Induced A1 Reactive Astrocytes in Brain in MPTP-Treated Murine Model of Parkinson’s Disease
Next Article in Special Issue
A Comprehensive Review of Emerging Trends and Innovative Therapies in Epilepsy Management
Previous Article in Journal
Resting State Dynamic Reconfiguration of Spatial Attention Cortical Networks and Visuospatial Functioning in Non-Verbal Learning Disability (NVLD): A HD-EEG Investigation
Previous Article in Special Issue
Responsive Neurostimulation of the Anterior Thalamic Nuclei in Refractory Genetic Generalized Epilepsy: A Case Series
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunological Imbalances Associated with Epileptic Seizures in Type 2 Diabetes Mellitus

by
Wendy N. Phoswa
* and
Kabelo Mokgalaboni
Department of Life and Consumer Sciences, University of South Africa (UNISA), Science Campus, Private Bag X6, Florida, Roodepoort 1710, South Africa
*
Author to whom correspondence should be addressed.
Brain Sci. 2023, 13(5), 732; https://doi.org/10.3390/brainsci13050732
Submission received: 27 March 2023 / Revised: 24 April 2023 / Accepted: 26 April 2023 / Published: 27 April 2023
(This article belongs to the Special Issue Etiology, Symptoms and Treatment of Epilepsy)

Abstract

:
Purpose of the review: Type 2 diabetes mellitus (T2DM) is a global health burden that leads to an increased morbidity and mortality rate arising from microvascular and macrovascular complications. Epilepsy leads to complications that cause psychological and physical distress to patients and carers. Although these conditions are characterized by inflammation, there seems to be a lack of studies that have evaluated inflammatory markers in the presence of both conditions (T2DM and epilepsy), especially in low-middle-income countries where T2DM is epidemic. Summary findings: In this review, we describe the role of immunity in the seizure generation of T2DM. Current evidence shows an increase in the levels of biomarkers such as interleukin (IL-1β, IL-6, and IL-8), tumour necrosis factor-α (TNF-α), high mobility group box-1 (HMGB1), and toll-like receptors (TLRs) in epileptic seizures and T2DM. However, there is limited evidence to show a correlation between inflammatory markers in the central and peripheral levels of epilepsy. Conclusions: Understanding the pathophysiological mechanism behind epileptic seizures in T2DM through an investigation of immunological imbalances might improve diagnosis and further counter the risks of developing complications. This might also assist in delivering safe and effective therapies to T2DM patients affected, thus reducing morbidity and mortality by preventing or reducing associated complications. Moreover, this review also provides an overview approach on inflammatory cytokines that can be targeted when developing alternative therapies, in case these conditions coexist.

1. Introduction

Diabetes mellitus (DM) is a chronic metabolic disorder characterised by hyperglycaemia caused by complete or partial insufficiency of insulin secretion and insulin action [1]. There are two types of diabetes mellitus, namely, type 1 (insulin-dependent) and type 2 (non-insulin-dependent), also known as adult-onset diabetes [2,3]. T2DM is the most common and accounts for 90–95% of all cases of DM [4,5]. It is predicted that the prevalence of T2DM will continue to increase in the next twenty years, and more than 70% of patients will come from developing countries, with the majority being around the ages of 45 and 64 years [6]. A recent report by the International Diabetes Federation has shown an alarming prevalence of DM, with 537 diabetic patients worldwide [7]. T2DM is multifactorial, resulting from alterations in genetic, environmental, and behavioural risk factors [8,9,10]. People with T2DM are more susceptible to various complications, often leading to premature death. Amongst these complications are different types of seizures that occur in about 25% of patients with DM [11,12,13,14]. The precise aetiology of seizures in diabetic patients remains unclear; however, it is speculated to result from physiological factors, such as oxidative stress, immune abnormalities, microvascular lesions in the brain, an impaired brain–blood barrier (BBB), metabolic factors, and genetic mutation [11,15,16,17,18] (Figure 1). To date, there are no international criteria for the diagnosis and treatment of this condition, thus making it difficult to diagnose and treat this condition in its early stages. Nevertheless, anti-seizure medications, such as levetiracetam, carbamazepine, lamotrigine, topiramate, and zonisamide, are commonly recommended, primarily to manage seizures in diabetic patients [19]. This can reduce secondary complications that might be associated with seizures, especially in diabetes. Furthermore, existing evidence shows that anti-diabetic medications can also be beneficial in DM patients with epileptic seizures, as they can normalise blood glucose levels and prevent a further epileptic attack when patients are no longer using such drugs [11]. Despite this evidence, it is still unclear how immunological indices and inflammatory pathways contribute to the aetiology of epilepsy in diabetic patients, especially in middle-low-income countries. Therefore, our current review discusses immunological, inflammatory imbalances, implications and genetic mutations associated with epileptic seizures in T2DM.

2. Pathophysiology of Epileptic Seizures

The pathophysiology of epilepsy involves the series of events that contributes to neural hyperexcitability. Some implicated mechanisms impair ion homeostasis, neurotransmitter uptake, and blood BBB [21,22,23]. The neurons in the brain interact with each other by releasing neurotransmitters through axons, thus sending neural messages. The neurotransmitters have excitatory or inhibitory effects. The action potential of a neuron depends solely on the balance between the neurotransmitters’ excitatory and inhibitory effects [24]. The increased release of excitatory glutamate upregulates N-methyl-D-aspartate (NMDA) receptors, resulting in the accumulation of calcium ions [25]. An excess of calcium ions activates neuronal nitric oxide synthase (nNOS), which increases the production of nitric oxide (NO) and neurological apoptosis due to deoxyribonucleic acid (DNA) damage associated with excessive free radicals, all of which contributes to the development and progression of epilepsy [26] (Figure 1).

3. Immunological Expression in Epilepsy

According to Vezzani et al. (2016), animal models of epilepsy are characterised by the presence of peripheral immune cells [27]. Lymphocytes, in particular, have been observed in the hippocampus following status epilepticus induced by systemic pilocarpine injection [28] or intrahippocampal administration of kainic acid in mice [29]. Active brain extravasation of these cells may alter BBB permeability [28,30]. The critical question is whether this phenomenon relates to tissue hyperexcitability or neuropathology. A study by Fabene and colleagues in 2008 revealed that epileptic seizures in an epileptic mouse model induced by pilocarpine injection upregulate the expression of vascular cell adhesion molecules (VCAM-1), concomitant to an increased leukocyte arrest in the brain [28]. This mechanism is mediated by the leukocyte mucin P-selectin glycoprotein ligand-1 (PSGL-1) and leukocyte integrins alpha-4-beta-1 [28]. This team further revealed that inhibition of contact between leukocytes and vasculature by blocking antibodies or by genetically manipulating PSGL-1 genes significantly reduces seizure attacks, with the potential of alleviating the onset of epilepsy [28].
In contrast, in intracerebral kainic-acid-treated mice, macrophages and T-cells prevent neutrophil extravasation, delay the onset, and thus reduce the recurrence of spontaneous seizures [29]. Epilepsy is a health burden without an identifiable cause [31,32]. The pathophysiological mechanism underlying this disease has been shown to involve both innate and adaptive immune responses, such as T- and B-cell activation, auto-AB production, and activation of inflammatory responses in the epileptogenic foci [33,34]. Immune cells activating inflammatory response have been observed in brain cells, such as microglia, astrocytes, and neurons, from patients with pharmacoresistant epileptic seizures [35]. In addition to an inflammatory response in epileptic seizures, cytokines also activate inflammation in epileptic seizures [36]. The inflammatory molecules observed in epileptic brain tissues are not only responsible for an inflammatory response per se; however, they function as neuromodulators by activating their receptors expressed by neurons, thus affecting neuronal function and excitability [37]. Therefore, it is speculated that the over-activation of inflammatory response in brain cells and the neuronal regions might contribute to the pathophysiological mechanism of seizure generation [38,39]. It is consequently critical that considerable research focus be placed on understanding the implication of immunological mechanisms in the aetiology of epilepsy. A better understanding of these mechanisms in epilepsy might be explored as a target for developing alternative therapies to epileptic seizures.

4. Inflammation in Epilepsy and Diabetes Mellitus

4.1. Central Inflammation in Epilepsy

The nuclear factor kappa-beta (NF-κβ) signalling system regulates inflammation and further contributes to inflammation observed in epilepsy [40,41,42]. NF-κβ initiates neuroinflammation through a series of mechanisms, including cyclooxygenase (COX-2), the mammalian target of rapamycin, and mitogen-activated protein kinase. These mechanisms upregulate the gene expression and activity of NF-κβ [43]. Additionally, HMGB1, NF-κβ, TNF-α, and IL-1 may, respectively, activate toll-like receptor 4 (TLR-4), TNF-receptor (TNFR), and IL-1R [44].
Evidence from animal models of epilepsy has shown an upregulated expression of COX-2 in hippocampus neurons after one hour of seizure; however, treatment with COX-2 inhibitors exhibited beneficial effects [45,46,47]. Prostaglandin (PGE2), a derivative of an enzyme, COX-2, contributes to the activation of prostaglandin 1, 2, 3, and 4 receptors (EP1, EP2, EP3, and EP4). This further promotes the release of calcium ions and mediates activities that impair neural function, including plasticity, neurologic deficiency, and hyperexcitability [48]. Interestingly, this reduces the threshold of seizure amongst animals and individuals with epilepsy [49,50]. Although inflammation is crucial to the aetiology of epilepsy, it is critical that future clinical investigations focus on the fundamental knowledge of inflammatory pathways that appear to promote and exacerbate epileptic complications.

4.2. Inflammation in T2DM

In hyperglycaemic conditions, insulin resistance results in inflammation. It has been revealed by several researchers that T2DM is associated with inflammation. Our group has previously shown an increased level of tumour necrosis factor alpha (TNF-α), interleukin-6 (IL-6) and interleukin-1-beta (IL-1β) in T2DM [51]. This suggests that increased inflammation is associated with cardiovascular diseases. Notably, the anti-inflammatory cytokine IL-4 is significantly decreased amongst T2DM patients [52]. T2DM is exacerbated by an imbalance between pro-inflammatory and anti-inflammatory cytokines.

5. Brain Cells as a Primary Site for the Activation of Inflammatory Responses in Epileptic Seizures

5.1. Glial Cells

Glial cells are reported to be the primary site for inflammatory molecule activation during epileptic seizures [36,53,54,55,56]. Upon their activation, they release several pro-inflammatory cytokines, such as IL-1β and TNF-α [36,57] (Figure 2). High-mobility group box-1 (HMGB-1) has been widely recognised as a biomarker of epilepsy [35,58]. It mediates the immune response by stimulating macrophage and endothelial cell activation, which triggers the production of TNF-α, IL-1, and IL-6 by binding to the receptor for advanced glycation end products (RAGE) and Toll-Like-receptors (TLR)-4. This further activates the nuclear factor kappa-light-chain-enhancer of activated β-cells (NF-κβ), subsequently increasing pro-inflammatory cytokine levels [59]. Evidence from preclinical studies shows that animal models exposed to lipopolysaccharide (LPS)-induced inflammatory response develop seizure due to pronounced levels of pro-inflammatory cytokines, such as IL-1β, TNF-α, and HMGB1, as seen in epilepsy comorbidities [57,60,61]. The accumulation of extracellular HMGB1, following apoptosis and highly elevated pro-inflammatory cytokine levels, promotes inflammation, which exacerbates the condition. Notably, these cytokines are partly elevated in both T2DM and epilepsy (Figure 2).

5.2. Microglia and Astrocytes

Animal models have shown other types of brain cells (microglia and astrocytes) that were affected during the pathophysiological mechanism of epileptic seizures [62]. It has been reported that activated microglia play a principal role in the production of cytokines that are also involved in the pathophysiology of epileptic seizures [63]. Microglia are called brain-resident immune cells partly because they can produce and release various cytokines [64]. A study by Benson et al. (2015) investigated the microglial expression of inflammatory cytokines using flow cytometry and quantitative real-time polymerase chain reaction (PCR) in a pilocarpine-induced SE. Their findings indicated that the microglia increased the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, Arg1, IL-4, and IL-10) [65]. Cytokine activation in the microglial cells of people with epilepsy has been shown to occur due to TLR signalling. Microglial cells respond to the TLR3 agonist polyinosinic and the TLR4 agonist lipopolysaccharide (LPS), thus promoting the production of inflammatory cytokines [66]. The TLR has also been implicated in the pathophysiology of diabetes mellitus. Kolek et al. (2004) showed that TRL4 [lipopolysaccharide (LPS) receptor] affects the innate immune response as well as the prevalence of T2D or metabolic syndrome and atherosclerosis [67]. TRL4 has also been reported to promote insulin resistance [68]. Insulin resistance contributes to the pathogenesis of DM, more particularly T2DM [69]. Interestingly, the findings by Wang et al. (2015) demonstrated the hyperglycaemia-induced overexpression and activation of TLR4 in endothelial cells. They further revealed that TLR4 leads to the activation of inflammatory responses that contribute to the pathogenesis of diabetic retinopathy [63]. More interestingly, Taha et al. (2018) suggested that high levels of TLR4 are associated with T2DM [70].
TLR4 plays a pivotal role in the brain and central nervous system (CNS) by regulating neuroinflammation [71]. The activation of TLR4 has a beneficial scavenging effect on amyloid beta (Aβ). However, the chronic activation of Aβ leads to Aβ deposition in the brain and has been widely associated with the pathogenesis of Alzheimer’s disease [72]. Alzheimer’s disease is a neurodegenerative disease that is associated with complications of diabetes mellitus [73]. TLR4 has also been reported to link diabetes mellitus and Alzheimer’s disease [74]. Therefore, we speculate that the TLR4 signalling pathway may also be a potential link between T2DM and epileptic seizures. TNF-α is released from activated microglia and astrocytes, and it can induce epilepsy. In a nutshell, TNF-α controls the synaptic function in astrocytes and regulates brain activity by increasing glutamate release, decreasing the production of gamma-aminobutyric acid, generating neuroinflammation [75]. Therefore, any therapeutic approach that can reduce TNF-α-associated glutamate may be of relevance in the search for an ideal epileptic treatment.
Microglial stimulation has been demonstrated to contribute to epilepsy through the HMGB1-TLR2/4-NF-kβ-mediated pathway. Of interest is the unique potential of this protein, HMGB1, as a treatment and non-invasive biomarker for epilepsy and patients at high risk of developing epilepsy [76]. It has been proven that the HMGB1 level rises after four hours of drug-resistant epilepsy seizure events, making HMGB1 a promising marker for epilepsy [77]. On the other hand, children with febrile seizures have increased serum levels of HMGB1 compared to normal children [78]. An increased level of HMGB1 induces inflammation, which may lead to additional complications associated with epilepsy. It is therefore critical to reduce inflammation in epileptic patients in order to reduce inflammatory-related secondary effects. Recently, the therapeutic strategy for reducing HMGB1 has been re-investigated, and the use of HMGB1 inhibitors has been well described in mouse models, with encouraging outcomes [79].

6. Peripheral Inflammatory Cytokines in Epilepsy

Brain inflammation is not the only source of the pathogenesis of epilepsy; peripheral inflammation also plays a critical role in the development of epilepsy [80]. Several findings have indicated that central-nervous-system inflammatory cytokines correlate with peripheral inflammation. A study by Chmielewska et al. (2021) observed increased plasma IL-1β and IL-6 in electrically induced hippocampal epilepsy in rats [81]. In agreement with this, Huang et al. (2018) also reported increased serum IL-2 and IL-6 in a rat model of seizures induced by intraperitoneal injection with kainic acid [82]. More recently, their findings were validated by Shin et al. (2022), who also observed increased serum IL-2 and IL-6 in tonic-clonic seizures [83].
Interestingly in epileptic patients, Basnyat et al. (2023) reported elevated IL-6 levels in the blood, which were associated with high antibodies against glutamic acid decarboxylase (GADA) titters [84]. These suggest that IL-6 can be used as a biomarker to understand the immunological pathways implicated in the pathophysiology of GADA-associated autoimmune epilepsy. HMGB1 is an inflammatory marker widely associated with epilepsy; however, currently, there is a paucity of evidence showing a correlation between central and peripheral HMGB1 levels. For example, Wang et al. (2021) determined the expression profiles of HMGB1 in cerebrospinal fluid (CSF) and paired serum [85]. They found that HMGB1 was only increased in the CSF, without a correlation between CSF and serum HMGB1 levels [85]. They further concluded that HMGB1 might be the main contributor to seizure mechanisms and that CSF HMGB1 can be used as a predictive marker in epilepsy [85]. Moreover, an understanding of the epileptic neurobiology of inflammation might be crucial to the identification of ideal markers that can be used as alternative therapeutic targets for the prevention and treatment of epileptic seizures.

7. Immunological Factors Commonly Contribute to the Pathogenesis of Diabetes

Immunological factors that commonly contribute to the pathogenesis of DM have been associated with two factors: (1) the activation of inflammasomes and (2) the release of pro-inflammatory cytokines in response to damage-associated molecular patterns (DAMPs). The inflammasome is a multiprotein complex needed for caspase-1 processing and activating inflammatory cytokines, such as IL-1β and IL-18 [86]. This activation of inflammatory cytokines occurs when damage-associated DAMPs, pathogen-associated molecular pattern molecules (PAMs, e.g., lipopolysaccharide), nucleotide-binding oligomerisation domain-like receptors (NLRs) or absent in melanoma 2 (AIM2) form a protein complex with pro-caspase-1 and the activation of TLR [86,87].
The aberrant activation of inflammasomes has been associated with the pathogenesis of autoimmune, autoinflammatory, chronically inflammatory, and metabolic diseases [88,89]. The NLR family activates abundant inflammasomes [90], which have been characterised by (1) the NLRP1/NALP1b inflammasome [91], (2) the NLRC4/IPAF inflammasome [92], (3) the NLRP3/NALP3 inflammasome [93], and (4) the AIM2-containing inflammasome [94]. Interestingly, NLRP3 inflammasomes are said to be activated by both exogenous (including DAMPs and MAPs) [95] and endogenous molecules, such as crystalline molecules, extracellular ATP, receptor P2X7 receptor (P2X7R) (through its cell surface), A fibrils, lipopolysaccharide (LPS), hyaluronan, and uric acid crystals [96].
Moreover, ATP receptors, such as P2X4, P2X7, P2Y4, P2Y6, P2Y7, and P2Y12, have been reported to be expressed by microglial cells [97,98]. Avignone et al. (2018) reported that P2Y6, P2Y4, P2Y6, P2Y7, and P2Y12 mRNA levels increased in the hippocampus in a kainic-acid-induced mouse model of mesial temporal lobe epilepsy (MTLE) after status epilepticus [99]. It is known that P2X7 activation promotes IL-1β processing and TNF-α expression, which are implicated in the pathophysiology of seizures [100]. However, regarding whether P2X7 has proconvulsive or anticonvulsive effects that vary according to the animal models of status epilepticus, P2X7 plays proconvulsive roles in pilocarpine-induced status epilepticus and anticonvulsive roles in status epilepticus triggered by intra-amygdala injection of kainic acid [101,102]. In addition to the role of NLRs, TLRs are implicated in the pathogenesis of T2DM and related complications [103,104,105,106]. Hence, this suggests that TLRs contribute to the disease’s development and to the pathogenesis of this condition [87]. It has been documented that TLR increases blood glucose levels, pro-inflammatory cytokines, and oxidative stress [107,108] (Figure 1).
There is increasing evidence linking HMGB1 with T2DM and obesity. A study conducted in China reported that increased plasma HMGB1 was associated with insulin resistance, whereas increased serum HMGB1 was linked to pro-inflammatory cytokines induced by T2DM and obesity [109]. Jeong et al. (2022) recently indicated that HMGB1 could exacerbate brain insulin resistance by reducing insulin receptor expression and deactivating the insulin signalling pathway. This can negatively impact the brain cells by preventing glucose transportation to these cells, thus leading to epileptic seizures [110]. In addition, T2DM can lead to oxidative stress, which causes BBB damage [111,112,113]. Damaged BBB can be vulnerable to neurological conditions such as epilepsy [114,115]. Notably, BBB leakage induces epilepsy [116] by increasing the production of glutamate. An increase in glutamate promotes excitatory activity in the brain, thus resulting in the development and exacerbation of epileptic seizure [117]. Recently, Chen and colleagues have reported the use of glutamate receptor antagonists as an effective treatment against epilepsy [117].
Other cytokines that are reportedly increased in T2DM include IL-6 [118,119,120,121,122], IL-2 [123,124], and TNF-α, which have been strongly associated with the pathogenesis of T2DM [125,126]. A vast trove of evidence from previous literature indicates similarities in immunological changes that occur in diabetes and epilepsy, which explains why T2DM patients are at an increased risk of being predisposed to epileptic seizures or epilepsy in the long run. In addition, both T2DM and epileptic aetiology seems to emanate from similar inflammatory pathways, as demonstrated by increased pro-inflammatory cytokines. This suggests an alternative approach that researchers can focus on when developing therapies against complications in both T2DM and epilepsy.

8. Conclusions

The aetiology of epilepsy is multifaceted; however, the imbalance in anti- and pro-inflammatory cytokines seems to implicate pathways in CNS and systemic tissue. The available evidence from preclinical and clinical studies shows similarities in the expression of immunological, pro-inflammatory and anti-inflammatory biomarkers, including IL-1β, IL-6, IL-8, HMGB1, TNF-α, TLRs, and PX27, implicated in the pathogenesis of T2DM and epileptic seizures. This suggests that there could be a close correlation between these two conditions. This creates a background that future research can focus on to further develop therapies against inflammatory conditions in instances where these two conditions coexist. Some of the approaches may include targeting HMGB1, which seems to exacerbate inflammation in epilepsy. Monitoring alterations in the BBB structure is also recommended in T2DM in order to prevent the pathogenesis of epilepsy. Despite this evidence, it is still crucial to conduct more clinical studies to ascertain these finding.

Author Contributions

Conceptualization and design, W.N.P.; methodology, W.N.P. and K.M.; validation, W.N.P. and K.M.; resources, W.N.P. and K.M.; writing—original draft preparation, W.N.P.; writing—review and editing, W.N.P. and K.M. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was required to conduct the research.

Institutional Review Board Statement

This article does not contain any studies with human participants or animals performed by any of the authors.

Informed Consent Statement

For this type of study, informed consent is not required.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vinay, K.; Abbas, A.K.; Fauston, N. Robbins and Cotran Pathologic Basis of Disease: Saunders; Elsevier: Beijing, China, 2005; Volume 8, pp. 208–221. [Google Scholar]
  2. Association, A.D. 2. Classification and diagnosis of diabetes: Standards of medical care in diabetes—2019. Diabetes Care 2019, 42 (Suppl. S1), S13–S28. [Google Scholar] [CrossRef] [PubMed]
  3. Mukhtar, Y.; Galalain, A.; Yunusa, U. A modern overview on diabetes mellitus: A chronic endocrine disorder. Eur. J. Biol. 2020, 5, 1–14. [Google Scholar] [CrossRef]
  4. Wu, Y.; Ding, Y.; Tanaka, Y.; Zhang, W. Risk factors contributing to type 2 diabetes and recent advances in the treatment and prevention. Int. J. Med. Sci. 2014, 11, 1185. [Google Scholar] [CrossRef] [PubMed]
  5. Olaniyi, K.S.; Amusa, O.A.; Ajadi, I.O.; Alabi, B.Y.; Agunbiade, T.B.; Ajadi, M.B. Repression of HDAC5 by acetate restores hypothalamic-pituitary-ovarian function in type 2 diabetes mellitus. Reprod. Toxicol. 2021, 106, 69–81. [Google Scholar] [CrossRef] [PubMed]
  6. Wild, S.H.; Roglic, G.; Green, A.; Sicree, R.; King, H. Global Prevalence of Diabetes: Estimates for the Year 2000 and Projections for 2030. Diabetes Care 2004, 27, 2569. [Google Scholar] [CrossRef]
  7. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Magliano, D.J. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res. Clin. Pract. 2022, 183, 109119. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, L.; Magliano, D.; Zimmet, P. The worldwide epidemiology of type 2 diabetes mellitus—Present and future perspectives. Nat. Rev. Endocrinol. 2012, 8, 228–236. [Google Scholar] [CrossRef] [PubMed]
  9. Sørensen, T.I.; Metz, S.; Kilpeläinen, T.O. Do gene–environment interactions have implications for the precision prevention of type 2 diabetes? Diabetologia 2022, 65, 1804–1813. [Google Scholar] [CrossRef]
  10. He, Y.; Lakhani, C.M.; Rasooly, D.; Manrai, A.K.; Tzoulaki, I.; Patel, C.J. Comparisons of polyexposure, polygenic, and clinical risk scores in risk prediction of type 2 diabetes. Diabetes Care 2021, 44, 935–943. [Google Scholar] [CrossRef]
  11. Yun, C.; Xuefeng, W. Association between seizures and diabetes mellitus: A comprehensive review of literature. Curr. Diabetes Rev. 2013, 9, 350–354. [Google Scholar] [CrossRef]
  12. Garrahy, A.; Thompson, C.J. Management of central diabetes insipidus. Best Pract. Res. Clin. Endocrinol. Metab. 2020, 34, 101385. [Google Scholar] [CrossRef]
  13. Yamada, Y.; Kitayama, K.; Oyachi, M.; Higuchi, S.; Kawakita, R.; Kanamori, Y.; Yorifuji, T. Nationwide survey of endogenous hyperinsulinemic hypoglycemia in Japan (2017–2018): Congenital hyperinsulinism, insulinoma, non-insulinoma pancreatogenous hypoglycemia syndrome and insulin autoimmune syndrome (Hirata’s disease). J. Diabetes Investig. 2020, 11, 554–563. [Google Scholar] [CrossRef] [PubMed]
  14. Taylor, T.; Holmes, S. Diabetes in Pediatric ED Patients. Pediatr. Emerg. Med. Rep. 2020, 25. Available online: https://www.reliasmedia.com/articles/145939-diabetes-in-pediatric-ed-patients (accessed on 26 March 2023).
  15. Youn, Y.; Sung, I.K.; Lee, I.G. The role of cytokines in seizures: Interleukin (IL)-1β, IL-1Ra, IL-8, and IL-10. Korean J. Pediatr. 2013, 56, 271. [Google Scholar] [CrossRef]
  16. Patel, M. Mitochondrial dysfunction and oxidative stress: Cause and consequence of epileptic seizures. Free Radic. Biol. Med. 2004, 37, 1951–1962. [Google Scholar] [CrossRef] [PubMed]
  17. Olowe, R.; Sandouka, S.; Saadi, A.; Shekh-Ahmad, T. Approaches for Reactive Oxygen Species and Oxidative Stress Quantification in Epilepsy. Antioxidants 2020, 9, 990. [Google Scholar] [CrossRef] [PubMed]
  18. Patel, M. Targeting Oxidative Stress in Central Nervous System Disorders. Trends Pharmacol. Sci. 2016, 37, 768–778. [Google Scholar] [CrossRef] [PubMed]
  19. Asadi-Pooya, A.A.; Sperling, M.R. Antiepileptic Drugs: A Clinician’s Manual; Oxford University Press: Oxford, UK, 2016. [Google Scholar]
  20. Wesół-Kucharska, D.; Rokicki, D.; Jezela-Stanek, A. Epilepsy in Mitochondrial Diseases—Current State of Knowledge on Aetiology and Treatment. Children 2021, 8, 532. [Google Scholar] [CrossRef]
  21. Löscher, W.; Friedman, A. Structural, Molecular, and Functional Alterations of the Blood-Brain Barrier during Epileptogenesis and Epilepsy: A Cause, Consequence, or Both? Int. J. Mol. Sci. 2020, 21, 591. [Google Scholar] [CrossRef]
  22. Zamay, T.N.; Zamay, G.S.; Shnayder, N.A.; Dmitrenko, D.V.; Zamay, S.S.; Yushchenko, V.; Kolovskaya, O.S.; Susevski, V.; Berezovski, M.V.; Kichkailo, A.S. Nucleic Acid Aptamers for Molecular Therapy of Epilepsy and Blood-Brain Barrier Damages. Mol. Ther. Nucleic Acids 2020, 19, 157–167. [Google Scholar] [CrossRef]
  23. Nikolic, L.; Nobili, P.; Shen, W.; Audinat, E. Role of astrocyte purinergic signaling in epilepsy. Glia 2020, 68, 1677–1691. [Google Scholar] [CrossRef] [PubMed]
  24. Mula, M. Emerging drugs for focal epilepsy. Expert Opin. Emerg. Drugs 2013, 18, 87–95. [Google Scholar] [CrossRef] [PubMed]
  25. Gong, P.; Zhang, S.; Ren, L.; Zhang, J.; Zhao, Y.; Mao, X.; Gan, L.; Wang, H.; Ma, C.; Lin, Y.; et al. Electroacupuncture of the trigeminal nerve causes N-methyl-D-aspartate receptors to mediate blood-brain barrier opening and induces neuronal excitatory changes. Front. Cell. Neurosci. 2022, 16, 1020644. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, S.D.; YW Chang, A.; Chuang, Y.C. The potential role of mitochondrial dysfunction in seizure-associated cell death in the hippocampus and epileptogenesis. J. Bioenerg. Biomembr. 2010, 42, 461–465. [Google Scholar] [CrossRef]
  27. Vezzani, A.; Fujinami, R.S.; White, H.S.; Preux, P.-M.; Blümcke, I.; Sander, J.W.; Löscher, W. Infections, inflammation and epilepsy. Acta Neuropathol. 2016, 131, 211–234. [Google Scholar] [CrossRef]
  28. Fabene, P.F.; Mora, G.N.; Martinello, M.; Rossi, B.; Merigo, F.; Ottoboni, L.; Bach, S.; Angiari, S.; Benati, D.; Chakir, A.; et al. A role for leukocyte-endothelial adhesion mechanisms in epilepsy. Nat. Med. 2008, 14, 1377–1383. [Google Scholar] [CrossRef] [PubMed]
  29. Zattoni, M.; Mura, M.L.; Deprez, F.; Schwendener, R.A.; Engelhardt, B.; Frei, K.; Fritschy, J.M. Brain infiltration of leukocytes contributes to the pathophysiology of temporal lobe epilepsy. J. Neurosci. 2011, 31, 4037–4050. [Google Scholar] [CrossRef] [PubMed]
  30. Librizzi, L.; Verde, D.V.; Colciaghi, F.; Deleo, F.; Regondi, M.C.; Costanza, M.; Cipelletti, B.; De Curtis, M. Peripheral blood mononuclear cell activation sustains seizure activity. Epilepsia 2021, 62, 1715–1728. [Google Scholar] [CrossRef] [PubMed]
  31. Paul, A.; Adeloye, D.; George-Carey, R.; Kolčić, I.; Grant, L.; Chan, K.Y. An estimate of the prevalence of epilepsy in Sub–Saharan Africa: A systematic analysis. J. Glob. Health 2012, 2, 020405. [Google Scholar] [CrossRef]
  32. Costa, C.; Vecchio, F.; Romoli, M.; Miraglia, F.; Cesarini, E.N.; Alù, F.; Calabresi, P.; Rossini, P.M. Cognitive Decline Risk Stratification in People with Late-Onset Epilepsy of Unknown Etiology: An Electroencephalographic Connectivity and Graph Theory Pilot Study. J. Alzheimers Dis. 2022, 88, 893–901. [Google Scholar] [CrossRef]
  33. Aronica, E.; Crino, P.B. Inflammation in epilepsy: Clinical observations. Epilepsia 2011, 52, 26–32. [Google Scholar] [CrossRef] [PubMed]
  34. Vezzani, A.; French, J.; Bartfai, T.; Baram, T.Z. The role of inflammation in epilepsy. Nat. Rev. Neurol. 2011, 7, 31–40. [Google Scholar] [CrossRef]
  35. Vezzani, A.; Lang, B.; Aronica, E. Immunity and inflammation in epilepsy. Cold Spring Harb. Perspect. Med. 2015, 6, a022699. [Google Scholar] [CrossRef] [PubMed]
  36. Aronica, E.; Ravizza, T.; Zurolo, E.; Vezzani, A. Astrocyte immune responses in epilepsy. Glia 2012, 60, 1258–1268. [Google Scholar] [CrossRef]
  37. Vezzani, A.; Maroso, M.; Balosso, S.; Sanchez, M.-A.; Bartfai, T. IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures. Brain Behav. Immun. 2011, 25, 1281–1289. [Google Scholar] [CrossRef]
  38. Mukhtar, I. Inflammatory and immune mechanisms underlying epileptogenesis and epilepsy: From pathogenesis to treatment target. Seizure 2020, 82, 65–79. [Google Scholar] [CrossRef] [PubMed]
  39. Semple, B.D.; Dill, L.K.; O’Brien, T.J. Immune challenges and seizures: How do early life insults influence epileptogenesis? Front. Pharmacol. 2020, 11, 2. [Google Scholar] [CrossRef]
  40. Vezzani, A.; Granata, T. Brain Inflammation in Epilepsy: Experimental and Clinical Evidence. Epilepsia 2005, 46, 1724–1743. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, M.; Xie, Y.; Shao, Y.; Chen, Y. LncRNA Snhg5 Attenuates Status Epilepticus Induced Inflammation through Regulating NF-κΒ Signaling Pathway. Biol. Pharm. Bull. 2022, 45, 86–93. [Google Scholar] [CrossRef] [PubMed]
  42. Xie, Y.; Wang, M.; Deng, X.; Chen, Y. Long non-coding RNA H19 alleviates hippocampal damage in convulsive status epilepticus rats through the nuclear factor-kappaB signaling pathway. Bioengineered 2022, 13, 12783–12793. [Google Scholar] [CrossRef]
  43. Cai, M.; Lin, W. The Function of NF-Kappa B During Epilepsy, a Potential Therapeutic Target. Front. Neurosci. 2022, 16, 851394. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, M.; Chen, Y. Inflammation: A Network in the Pathogenesis of Status Epilepticus. Front. Mol. Neurosci. 2018, 11, 341. [Google Scholar] [CrossRef]
  45. Rojas, A.; Jiang, J.; Ganesh, T.; Yang, M.-S.; Lelutiu, N.; Gueorguieva, P.; Dingledine, R. Cyclooxygenase-2 in epilepsy. Epilepsia 2014, 55, 17–25. [Google Scholar] [CrossRef]
  46. Dhir, A. An update of cyclooxygenase (COX)-inhibitors in epilepsy disorders. Expert Opin. Investig. Drugs 2019, 28, 191–205. [Google Scholar] [CrossRef] [PubMed]
  47. Okada, K.; Yuhi, T.; Tsuji, S.; Yamashita, U. Cyclooxygenase-2 expression in the hippocampus of genetically epilepsy susceptible El mice was increased after seizure. Brain Res. 2001, 894, 332–335. [Google Scholar] [CrossRef] [PubMed]
  48. Figueiredo-Pereira, M.E.; Rockwell, P.; Schmidt-Glenewinkel, T.; Serrano, P. Neuroinflammation and J2 prostaglandins: Linking impairment of the ubiquitin-proteasome pathway and mitochondria to neurodegeneration. Front. Mol. Neurosci. 2015, 7, 104. [Google Scholar] [CrossRef] [PubMed]
  49. Rawat, C.; Kukal, S.; Dahiya, U.R.; Kukreti, R. Cyclooxygenase-2 (COX-2) inhibitors: Future therapeutic strategies for epilepsy management. J. Neuroinflammation 2019, 16, 197. [Google Scholar] [CrossRef] [PubMed]
  50. Mesdaghinia, A.; Khazaee, P.; Heydari, A. The effect of cyclooxygenase-2 inhibition on pentylenetetrazole-induced seizure threshold in mice. KAUMS J. (FEYZ) 2018, 22, 258–266. [Google Scholar]
  51. Mokgalaboni, K.; Dludla, P.V.; Nyambuya, T.M.; Yakobi, S.H.; Mxinwa, V.; Nkambule, B.B. Monocyte-mediated inflammation and cardiovascular risk factors in type 2 diabetes mellitus: A systematic review and meta-analysis of pre-clinical and clinical studies. JRSM Cardiovasc. Dis. 2020, 9, 2048004019900748. [Google Scholar] [CrossRef]
  52. Araújo, L.S.; da Silva, M.V.; da Silva, C.A.; Borges, M.D.F.; Palhares, H.M.D.C.; Rocha, L.P.; Machado, J.R. Analysis of serum inflammatory mediators in type 2 diabetic patients and their influence on renal function. PLoS ONE 2020, 15, e0229765. [Google Scholar] [CrossRef]
  53. Devinsky, O.; Vezzani, A.; Najjar, S.; De Lanerolle, N.C.; Rogawski, M.A. Glia and epilepsy: Excitability and inflammation. Trends Neurosci. 2013, 36, 174–184. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, G.; Wang, J.; Xin, C.; Xiao, J.; Liang, J.; Wu, X. Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review). Mol. Med. Rep. 2021, 24, 493. [Google Scholar] [CrossRef] [PubMed]
  55. Han, C.; Liu, Y.; Guo, C.; Du, T.; Jiang, Y.; Wang, K.; Shao, X.; Meng, F.; Zhang, J. The lncRNA H19 binding to let-7b promotes hippocampal glial cell activation and epileptic seizures by targeting Stat3 in a rat model of temporal lobe epilepsy. Cell Prolif. 2020, 53, e12856. [Google Scholar] [CrossRef] [PubMed]
  56. Li, J.; Shi, D.; Wang, L.; Wu, G. Chronic neuroinflammation regulates cAMP response element-binding protein in the formation of drug-resistant epilepsy by activating glial cells. J. Neurorestoratology 2022, 10, 100006. [Google Scholar] [CrossRef]
  57. Vezzani, A.; Aronica, E.; Mazarati, A.; Pittman, Q. Epilepsy and brain inflammation. Exp. Neurol. 2011, 244, 11–21. [Google Scholar] [CrossRef] [PubMed]
  58. Raoof, R.M.; Al-Hamdany, M.; Noel, K.I. Role of High Mobility Group Box-1 in Status Epilepticus, From Pathophysiology to Biomarker and Therapeutic Potential. Ann. Coll. Med. Mosul 2022, 44, 37–41. [Google Scholar] [CrossRef]
  59. Gao, H.M.; Zhou, H.; Zhang, F.; Wilson, B.C.; Kam, W.; Hong, J.S. HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J. Neurosci. 2011, 31, 1081–1092. [Google Scholar] [CrossRef]
  60. Galic, M.A.; Riazi, K.; Pittman, Q.J. Cytokines and brain excitability. Front. Neuroendocr. 2012, 33, 116–125. [Google Scholar] [CrossRef]
  61. Pineda, E.; Shin, D.; You, S.J.; Auvin, S.; Sankar, R.; Mazarati, A. Maternal immune activation promotes hippocampal kindling epileptogenesis in mice. Ann. Neurol. 2013, 74, 11–19. [Google Scholar] [CrossRef] [PubMed]
  62. Hiragi, T.; Ikegaya, Y.; Koyama, R. Microglia after Seizures and in Epilepsy. Cells 2018, 7, 26. [Google Scholar] [CrossRef]
  63. Wang, W.-Y.; Tan, M.-S.; Yu, J.-T.; Tan, L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015, 3, 136. [Google Scholar] [PubMed]
  64. Andoh, M.; Ikegaya, Y.; Koyama, R. Synaptic Pruning by Microglia in Epilepsy. J. Clin. Med. 2019, 8, 2170. [Google Scholar] [CrossRef]
  65. Benson, M.J.; Manzanero, S.; Borges, K. Complex alterations in microglial M1/M2 markers during the development of epilepsy in two mouse models. Epilepsia 2015, 56, 895–905. [Google Scholar] [CrossRef] [PubMed]
  66. Olson, J.K.; Miller, S.D. Microglia Initiate Central Nervous System Innate and Adaptive Immune Responses through Multiple TLRs. J. Immunol. 2004, 173, 3916–3924. [Google Scholar] [CrossRef]
  67. Kolek, M.J.; Carlquist, J.F.; Muhlestein, J.B.; Whiting, B.M.; Horne, B.D.; Bair, T.L.; Anderson, J.L. Toll–like receptor 4 gene Asp299Gly polymorphism is associated with reductions in vascular inflammation, angiographic coronary artery disease, and clinical diabetes. Am. Heart J. 2004, 148, 1034–1040. [Google Scholar] [CrossRef]
  68. Kim, J.J.; Sears, D.D. TLR4 and insulin resistance. Gastroenterol. Res. Pract. 2010, 2010, 212563. [Google Scholar] [CrossRef] [PubMed]
  69. Samuel, V.T.; Shulman, G.I. The pathogenesis of insulin resistance: Integrating signaling pathways and substrate flux. J. Clin. Investig. 2016, 126, 12–22. [Google Scholar] [CrossRef] [PubMed]
  70. Taha, I.M.; Allah, A.M.A.; El Gayed, E.M.A. Expression of toll-like receptor 4 and its connection with type 2 diabetes mellitus. Cell. Mol. Biol. 2018, 64, 15–20. [Google Scholar] [CrossRef]
  71. Fiebich, B.L.; Batista, C.R.A.; Saliba, S.W.; Yousif, N.M.; De Oliveira, A.C.P. Role of Microglia TLRs in Neurodegeneration. Front. Cell. Neurosci. 2018, 12, 329. [Google Scholar] [CrossRef]
  72. Chen, Y.G. Research progress in the pathogenesis of Alzheimer’s disease. Chin. Med. J. 2018, 131, 1618–1624. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, H.J.; Seo, H.I.; Cha, H.Y.; Yang, Y.J.; Kwon, S.H.; Yang, S.J. Diabetes and Alzheimer’s disease: Mechanisms and nutritional aspects. Clin. Nutr. Res. 2018, 7, 229–240. [Google Scholar] [CrossRef]
  74. Huang, N.-Q.; Jin, H.; Zhou, S.-Y.; Shi, J.-S.; Jin, F. TLR4 is a link between diabetes and Alzheimer’s disease. Behav. Brain Res. 2017, 316, 234–244. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, R.; Xue, G.; Hölscher, C. The role of the TNFα-mediated astrocyte signaling pathway in epilepsy. Acta Epileptol. 2021, 3, 24. [Google Scholar] [CrossRef]
  76. Shi, Y.; Zhang, L.; Teng, J.; Miao, W. HMGB1 mediates microglia activation via the TLR4/NF-κB pathway in coriaria lactone induced epilepsy. Mol. Med. Rep. 2018, 17, 5125–5131. [Google Scholar] [CrossRef] [PubMed]
  77. Zhu, M.; Chen, J.; Guo, H.; Ding, L.; Zhang, Y.; Xu, Y. High Mobility Group Protein B1 (HMGB1) and Interleukin-1β as Prognostic Biomarkers of Epilepsy in Children. J. Child Neurol. 2018, 33, 909–917. [Google Scholar] [CrossRef] [PubMed]
  78. Choi, J.; Min, H.J.; Shin, J.-S. Increased levels of HMGB1 and pro-inflammatory cytokines in children with febrile seizures. J. Neuroinflammation 2011, 8, 135. [Google Scholar] [CrossRef] [PubMed]
  79. Yang, H.; Wang, H.; Andersson, U. Targeting Inflammation Driven by HMGB1. Front. Immunol. 2020, 11, 484. [Google Scholar] [CrossRef] [PubMed]
  80. Zabielski, P.; Chacinska, M.; Charkiewicz, K.; Baranowski, M.; Gorski, J.; Blachnio-Zabielska, A.U. Effect of metformin on bioactive lipid metabolism in insulin-resistant muscle. J. Endocrinol. 2017, 233, 329–340. [Google Scholar] [CrossRef]
  81. Chmielewska, N.; Maciejak, P.; Osuch, B.; Kursa, M.B.; Szyndler, J. Pro-inflammatory cytokines, but not brain- and extracellular matrix-derived proteins, are increased in the plasma following electrically induced kindling of seizures. Pharmacol. Rep. 2021, 73, 506–515. [Google Scholar] [CrossRef]
  82. Huang, W.-S.; Zhu, L. MiR-134 expression and changes in inflammatory cytokines of rats with epileptic seizures. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3479–3484. [Google Scholar]
  83. Shin, H.-R.; Chu, K.; Lee, W.-J.; Lee, H.S.; Kim, E.Y.; Son, H.; Moon, J.; Kim, N.; Jung, K.-Y.; Jung, K.-H.; et al. Neuropsychiatric symptoms and seizure related with serum cytokine in epilepsy patients. Sci. Rep. 2022, 12, 7138. [Google Scholar] [CrossRef]
  84. Basnyat, P.; Peltola, M.; Raitanen, J.; Liimatainen, S.; Rainesalo, S.; Pesu, M.; Peltola, J. Elevated IL-6 plasma levels are associated with GAD antibodies-associated autoimmune epilepsy. Front. Cell. Neurosci. 2023, 17, 1129907. [Google Scholar] [CrossRef]
  85. Wang, N.; Liu, H.; Ma, B.; Zhao, T.; Chen, Y.; Yang, Y.; Zhao, P.; Han, X. CSF high-mobility group box 1 is associated with drug-resistance and symptomatic etiology in adult patients with epilepsy. Epilepsy Res. 2021, 177, 106767. [Google Scholar] [CrossRef]
  86. Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef] [PubMed]
  87. Sepehri, Z.; Kiani, Z.; Nasiri, A.A.; Kohan, F. Toll-like receptor 2 and type 2 diabetes. Cell. Mol. Biol. Lett. 2016, 21, 2. [Google Scholar] [CrossRef] [PubMed]
  88. Franchi, L.; Muñoz-Planillo, R.; Núñez, G. Sensing and reacting to microbes through the inflammasomes. Nat. Immunol. 2012, 13, 325–332. [Google Scholar] [CrossRef] [PubMed]
  89. Menu, P.; Vince, J.E. The NLRP3 inflammasome in health and disease: The good, the bad and the ugly. Clin. Exp. Immunol. 2011, 166, 1–15. [Google Scholar] [CrossRef] [PubMed]
  90. Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef]
  91. Boyden, E.D.; Dietrich, W.F. Nalp1b controls mouse macrophage susceptibility to anthrax lethal toxin. Nat. Genet. 2006, 38, 240–244. [Google Scholar] [CrossRef]
  92. Zhao, Y.; Yang, J.; Shi, J.; Gong, Y.-N.; Lu, Q.; Xu, H.; Liu, L.; Shao, F. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature 2011, 477, 596–600. [Google Scholar] [CrossRef] [PubMed]
  93. Martinon, F.; Pétrilli, V.; Mayor, A.; Tardivel, A.; Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006, 440, 237–241. [Google Scholar] [CrossRef]
  94. Fernandes-Alnemri, T.; Yu, J.W.; Datta, P.; Wu, J.; Alnemri, E.S. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 2009, 458, 509–513. [Google Scholar] [CrossRef] [PubMed]
  95. Pressman, B.C. Biological Applications of Ionophores. Annu. Rev. Biochem. 1976, 45, 501–530. [Google Scholar] [CrossRef] [PubMed]
  96. Guo, H.; Callaway, J.B.; Ting, J.P.-Y. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015, 21, 677–687. [Google Scholar] [CrossRef]
  97. Puerto, A.D.; Wandosell, F.; Garrido, J.J. Neuronal and glial purinergic receptors functions in neuron development and brain disease. Front. Cell. Neurosci. 2013, 7, 197. [Google Scholar] [CrossRef] [PubMed]
  98. Verkhratsky, A.; Pankratov, Y.; Lalo, U.; Nedergaard, M. P2X receptors in neuroglia. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 151–161. [Google Scholar] [CrossRef]
  99. Avignone, E.; Ulmann, L.; Levavasseur, F.; Rassendren, F.; Audinat, E. Status Epilepticus Induces a Particular Microglial Activation State Characterized by Enhanced Purinergic Signaling. J. Neurosci. 2008, 28, 9133–9144. [Google Scholar] [CrossRef]
  100. Rappold, P.; Lynd-Balta, E.; Joseph, S. P2X7 receptor immunoreactive profile confined to resting and activated microglia in the epileptic brain. Brain Res. 2006, 1089, 171–178. [Google Scholar] [CrossRef] [PubMed]
  101. Jimenez-Pacheco, A.; Diaz-Hernandez, M.; Arribas-Blázquez, M.; Sanz-Rodriguez, A.; Olivos-Oré, L.A.; Artalejo, A.R.; Henshall, D.C. Transient P2X7 receptor antagonism produces lasting reductions in spontaneous seizures and gliosis in experimental temporal lobe epilepsy. J. Neurosci. 2016, 36, 5920–5932. [Google Scholar] [CrossRef]
  102. Kim, J.-E.; Kang, T.-C. The P2X7 receptor–pannexin-1 complex decreases muscarinic acetylcholine receptor–mediated seizure susceptibility in mice. J. Clin. Investig. 2011, 121, 2037–2047. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, P.; Zhang, Z.; Li, Y. Relevance of the Pyroptosis-Related Inflammasome Pathway in the Pathogenesis of Diabetic Kidney Disease. Front. Immunol. 2021, 12, 603416. [Google Scholar] [CrossRef] [PubMed]
  104. Zheng, Y.; Gou, X.; Zhang, L.; Gao, H.; Wei, Y.; Yu, X.; Pang, B.; Tian, J.; Tong, X.; Li, M. Interactions Between Gut Microbiota, Host, and Herbal Medicines: A Review of New Insights Into the Pathogenesis and Treatment of Type 2 Diabetes. Front. Cell. Infect. Microbiol. 2020, 10, 360. [Google Scholar] [CrossRef]
  105. Koushki, K.; Shahbaz, S.K.; Mashayekhi, K.; Sadeghi, M.; Zayeri, Z.D.; Taba, M.Y.; Sahebkar, A. Anti-inflammatory action of statins in cardiovascular disease: The role of inflammasome and toll-like receptor pathways. Clin. Rev. Allergy Immunol. 2021, 60, 175–199. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, J.; Zhang, H.; Su, Y.; Zhang, B. Application and prospect of targeting innate immune sensors in the treatment of autoimmune diseases. Cell Biosci. 2022, 12, 68. [Google Scholar] [CrossRef]
  107. Dasu, M.R.; Ramirez, S.; Isseroff, R.R. Toll-like receptors and diabetes: A therapeutic perspective. Clin. Sci. 2012, 122, 203–214. [Google Scholar] [CrossRef]
  108. Liu, T.; Zheng, W.; Wang, L.; Wang, L.; Zhang, Y. TLR4/NF-κB signaling pathway participates in the protective effects of apocynin on g0estational diabetes mellitus induced placental oxidative stress and inflammation. Reprod. Sci. 2020, 27, 722–730. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, H.; Qu, H.; Deng, H. Plasma HMGB-1 Levels in Subjects with Obesity and Type 2 Diabetes: A Cross-Sectional Study in China. PLoS ONE 2015, 10, e0136564. [Google Scholar] [CrossRef] [PubMed]
  110. Jeong, J.H.; Lee, D.H.; Song, J. HMGB1 signaling pathway in diabetes-related dementia: Blood-brain barrier breakdown, brain insulin resistance, and Aβ accumulation. Biomed. Pharmacother. 2022, 150, 112933. [Google Scholar] [CrossRef] [PubMed]
  111. Dokken, B.B. The Pathophysiology of Cardiovascular Disease and Diabetes: Beyond Blood Pressure and Lipids. Diabetes Spectr. 2008, 21, 160–165. [Google Scholar] [CrossRef]
  112. Banks, W.A.; Rhea, E.M. The Blood–Brain Barrier, Oxidative Stress, and Insulin Resistance. Antioxidants 2021, 10, 1695. [Google Scholar] [CrossRef]
  113. Choi, S.E.; Roy, B.; Freeby, M.; Mullur, R.; Woo, M.A.; Kumar, R. Prefrontal cortex brain damage and glycemic control in patients with type 2 diabetes. J. Diabetes 2020, 12, 465–473. [Google Scholar] [CrossRef] [PubMed]
  114. Mendes, N.F.; Pansani, A.P.; Carmanhães, E.R.F.; Tange, P.; Meireles, J.V.; Ochikubo, M.; Le Sueur-Maluf, L. The blood-brain barrier breakdown during acute phase of the pilocarpine model of epilepsy is dynamic and time-dependent. Front. Neurol. 2019, 10, 382. [Google Scholar] [CrossRef]
  115. Van Vliet, E.A.; Aronica, E.; Gorter, J.A. Blood–Brain Barrier Dysfunction, Seizures and Epilepsy. In Seminars in Cell Developmental Biology; Academic Press: Cambridge, MA, USA, 2015; Volume 38, pp. 26–34. [Google Scholar]
  116. Löscher, W. Epilepsy and Alterations of the Blood–Brain Barrier: Cause or Consequence of Epileptic Seizures or Both? In Physiology, Pharmacology and Pathology of the Blood-Brain Barrier; Cader, Z., Neuhaus, W., Eds.; Springer International Publishing: Berlin/Heidelberg, Germany; Cham, Switzerland, 2022; pp. 331–350. [Google Scholar]
  117. Chen, T.-S.; Huang, T.-H.; Lai, M.-C.; Huang, C.-W. The Role of Glutamate Receptors in Epilepsy. Biomedicines 2023, 11, 783. [Google Scholar] [CrossRef] [PubMed]
  118. Pradhan, A.D.; Manson, J.E.; Rifai, N.; Buring, J.E.; Ridker, P.M. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 2001, 286, 327–334. [Google Scholar] [CrossRef]
  119. Laishram, V.; Debnath, S.; Hijam, D.; Das, T.; Saha, S. Evaluation of serum interleukin 6 in type II diabetes mellitus. Int. J. Med. Res. Prof. 2017, 3, 156–160. [Google Scholar]
  120. Thamer, M.M.; Wong, C.F.; Ghali, Z.H. Estimation of Interleukin-4 (IL-4) and Interleukin-6 (IL-6) Levels in Sera From Patients with Type 2 Diabetes Mellitus. Indian J. Forensic Med. Toxicol. 2021, 15, 1732–1739. [Google Scholar]
  121. Chanda, D.; Ray, S.; Chakraborti, D.; Sen, S.; Mitra, A. Interleukin-6 Levels in Patients With Diabetic Polyneuropathy. Cureus 2022, 14, 21952. [Google Scholar] [CrossRef] [PubMed]
  122. Phosat, C.; Panprathip, P.; Chumpathat, N.; Prangthip, P.; Chantratita, N.; Soonthornworasiri, N.; Puduang, S.; Kwanbunjan, K. Elevated C-reactive protein, interleukin 6, tumor necrosis factor alpha and glycemic load associated with type 2 diabetes mellitus in rural Thais: A cross-sectional study. BMC Endocr. Disord. 2017, 17, 44. [Google Scholar] [CrossRef] [PubMed]
  123. Bosek, I.; Kuczerowski, R.; Miłek, T.; Rabijewski, M.; Kaleta, B.; Kniotek, M.; Piątkiewicz, P. The levels of interleukin-2 and interleukin-10 in patients with type 2 diabetes and colon cancer. Clin. Diabetol. 2018, 7, 114–121. [Google Scholar] [CrossRef]
  124. Suri, S.; Mitra, P.; Abhilasha, A.; Saxena, I.; Garg, M.K.; Bohra, G.K.; Sharma, P. Role of interleukin-2 and interleukin-18 in newly diagnosed type 2 diabetes mellitus. J. Basic Clin. Physiol. Pharmacol. 2021, 33, 185–190. [Google Scholar] [CrossRef] [PubMed]
  125. AlZamil, H. Elevated Serum TNF-α Is Related to Obesity in Type 2 Diabetes Mellitus and Is Associated with Glycemic Control and Insulin Resistance. J. Obes. 2020, 2020, 5076858. [Google Scholar] [CrossRef] [PubMed]
  126. Swaroop, J.J.; Naidu, J.; Rajarajeswari, D. Association of TNF-α with insulin resistance in type 2 diabetes mellitus. Indian J. Med. Res. 2012, 135, 127–130. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Brief mechanism of the pathophysiology of epilepsy. Different pathways can induce this; for example, oxidative stress due to an increased circulating reactive oxygen species (ROS), which results in imbalanced inflammatory cytokines. The latter stimulates inflammation and subsequently leads to neuroinflammation. Similarly, ROS can impair calcium ion metabolism, resulting in neural hyperexcitability. The loss of mitochondrial adenosine-triphosphate (ATP) has also been implicated in the pathogenesis of epilepsy. This seems to occur in three different pathways; for instance, the alteration of neurotransmitters due to the lack of ATP can induce neural hyperexcitability by increasing glutamate and the reduction of gamma-aminobutyric acid (GABA) in a mitochondrion. Secondly, less ATP impairs the sodium and potassium ion channels, resulting in nerve depolarisation. On the other hand, the impaired brain–blood barrier (BBB) also triggers neuroinflammation, causing neural degeneration. Altogether, these mechanisms cause neuronal degeneration, which facilitates the pathogenicity of epilepsy [20].
Figure 1. Brief mechanism of the pathophysiology of epilepsy. Different pathways can induce this; for example, oxidative stress due to an increased circulating reactive oxygen species (ROS), which results in imbalanced inflammatory cytokines. The latter stimulates inflammation and subsequently leads to neuroinflammation. Similarly, ROS can impair calcium ion metabolism, resulting in neural hyperexcitability. The loss of mitochondrial adenosine-triphosphate (ATP) has also been implicated in the pathogenesis of epilepsy. This seems to occur in three different pathways; for instance, the alteration of neurotransmitters due to the lack of ATP can induce neural hyperexcitability by increasing glutamate and the reduction of gamma-aminobutyric acid (GABA) in a mitochondrion. Secondly, less ATP impairs the sodium and potassium ion channels, resulting in nerve depolarisation. On the other hand, the impaired brain–blood barrier (BBB) also triggers neuroinflammation, causing neural degeneration. Altogether, these mechanisms cause neuronal degeneration, which facilitates the pathogenicity of epilepsy [20].
Brainsci 13 00732 g001
Figure 2. Demonstration of the level of circulating pro-inflammatory cytokines in epileptic seizures and T2DM. In both conditions, there is an increased level of pro-inflammatory cytokines, implying that there could be a direct link or inflammatory relationship between them. TNF-α, tumour necrosis factor-alpha; IL, interleukin; CRP, C-Reactive Protein; HMGB1, High-mobility group box-1; TLRs, toll-like receptors.
Figure 2. Demonstration of the level of circulating pro-inflammatory cytokines in epileptic seizures and T2DM. In both conditions, there is an increased level of pro-inflammatory cytokines, implying that there could be a direct link or inflammatory relationship between them. TNF-α, tumour necrosis factor-alpha; IL, interleukin; CRP, C-Reactive Protein; HMGB1, High-mobility group box-1; TLRs, toll-like receptors.
Brainsci 13 00732 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Phoswa, W.N.; Mokgalaboni, K. Immunological Imbalances Associated with Epileptic Seizures in Type 2 Diabetes Mellitus. Brain Sci. 2023, 13, 732. https://doi.org/10.3390/brainsci13050732

AMA Style

Phoswa WN, Mokgalaboni K. Immunological Imbalances Associated with Epileptic Seizures in Type 2 Diabetes Mellitus. Brain Sciences. 2023; 13(5):732. https://doi.org/10.3390/brainsci13050732

Chicago/Turabian Style

Phoswa, Wendy N., and Kabelo Mokgalaboni. 2023. "Immunological Imbalances Associated with Epileptic Seizures in Type 2 Diabetes Mellitus" Brain Sciences 13, no. 5: 732. https://doi.org/10.3390/brainsci13050732

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop