Back to Journals » Drug Design, Development and Therapy » Volume 16

Protective Effects of Pioglitazone on Cognitive Impairment and the Underlying Mechanisms: A Review of Literature

Authors Alhowail A , Alsikhan R , Alsaud M, Aldubayan M, Rabbani SI

Received 20 March 2022

Accepted for publication 23 August 2022

Published 31 August 2022 Volume 2022:16 Pages 2919—2931

DOI https://doi.org/10.2147/DDDT.S367229

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Prof. Dr. Tin Wui Wong



Ahmad Alhowail,1 Rawan Alsikhan,1,2 May Alsaud,1 Maha Aldubayan,1 Syed Imam Rabbani1

1Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia; 2Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Unaizah, Al Qassim, 51911, Kingdom of Saudi Arabia

Correspondence: Ahmad Alhowail, Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia, Tel +9665672025858, Email [email protected]

Abstract: Pioglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) agonist, is known to have anti-inflammatory and anti-oxidant effects on the brain, and its clinical potential in the treatment of cognitive impairment in diseases such as Alzheimer’s disease (AD) and Parkinson disease (PD) is currently being explored. This review focused on the reported beneficial effects of pioglitazone on cognitive dysfunction and summarized the associated mechanisms associated with pioglitazone-induced improvement in cognitive dysfunction. Our review of the relevant literature indicated that there is conclusive evidence of the effect of pioglitazone on improving cognitive impairment via its agonistic effect on PPAR-γ. Further, several mechanisms of action have been reported, and these include enhanced NF-kB and p38 activity; regulation of the pro-inflammatory cytokines IL-1, IL-6, and TNF-α; inhibition of Aβ production; alterations in the levels of mitochondrial proteins such as mitoNEET; regulation of protein kinases such as CDK5 and JNK; regulation of ROS and MDA levels and the levels of the antioxidant proteins TRX1 and PON2; and increased expression of thyroid hormone receptors. Despite these promising findings, pioglitazone treatment is also associated with cardiovascular risks, such as weight gain and edema, which subsequently increase the risk of mortality. Further, it has been documented that pioglitazone may be unable to cross the blood–brain barrier when administered in certain forms, and it can also cause cell death when administered at high concentrations. Therefore, further research is required to explore the effects of acute and chronic pioglitazone treatment on memory function and the associated risks, in order to determine its clinical applicability in the treatment of cognitive disorders. Nonetheless, the current literature does demonstrate that pioglitazone promotes the function of PPAR receptors in ameliorating inflammation, oxidative stress, amyloidogenesis, and hypothyroidism, and enhancing neurogenesis, synaptic plasticity, and mitochondrial function. Therefore, these mechanisms of PPAR receptors warrant further investigation in order to establish the clinical applicability of pioglitazone in the treatment of cognitive disorders, such as PD and AD, and neuronal impairment in conditions such as diabetes.

Keywords: peroxisome proliferator-activated receptor gamma agonist, cognitive impairment, neuroinflammation, mitochondria

Introduction

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily and are comprised of three isoforms: PPAR-α, PPAR-β, and PPAR-γ. PPAR-γ is considered to be a regulator of glucose metabolism and fatty acid synthesis via the activation of factors that promote adipocyte differentiation.1 PPAR-γ is also known to be involved in the regulation of several physiological processes, such as the increased response of insulin receptors to insulin, lipid metabolism, and cellular proliferation.2 Therefore, PPAR-γ is an interesting therapeutic target for the treatment of metabolic disorders.

Table 1 Effect of Pioglitazone on Different Organ System

Table 2 Subtypes of PPAR Receptors and Their Distribution

Pioglitazone is a thiazolidinedione that acts as an activator of PPAR-γ and is commonly used to treat hypoglycemia. Recent studies have shown that certain pioglitazone formulations can cross the blood–brain barrier and improve the response of insulin receptors to insulin, regulate glucose metabolism in the brain, and reduce neuroinflammation.3,4 Attenuation of neuroinflammation can improve cognitive impairment. Accordingly, pioglitazone administration has been shown to lead to improvement in cognitive impairment caused by Alzheimer's disease (AD) and Parkinson disease (PD), as well as improvement in dopaminergic neuronal survival in the brain.5–11 However, although the results of preclinical studies on model animals and cells lines have been promising, there is not enough evidence to conclusively support the application of pioglitazone for the treatment of cognitive disorders in the clinical setting. In order to understand its clinical potential, it is important to further explore its mechanisms of action that affect cognitive function.

With regard to its mechanism of action in improving cognitive impairment, pioglitazone has been shown to ameliorate inflammation, oxidative stress, amyloidogenesis, and hypothyroidism, as well as enhance neurogenesis, synaptic plasticity, and mitochondrial function via its effects on the relevant PPAR-mediated pathways. Some of these effects of pioglitazone include promoting the anti-inflammatory NF-kB and p38 pathways; inhibiting the expression of beta-secretase 1, which is important for Aβ production; enhancing the expression of proteins that are important for mitochondrial function, such as PGC-1α and mitoNEET; inhibiting the increase in the production of the pro-inflammatory cytokines IL-1, IL-6, and TNF-α induced by bacterial lipopolysaccharides; regulation of the expression of protein kinases such as AKT and JNK; modulation of the antioxidant proteins TRX1, BDNF, and calcium–calmodulin-dependent protein kinase II (CaMKII); inhibition of ROS and MDA production; promotion of thyroid function via increased activation of thyroid hormone receptors; and modulation of the LRP1, GSK-3β, and Nrf2/ARE pathways, which are involved in synaptic transmission (Table 1). Despite these promising findings, PPAR-γ activation has been reported to increase cardiovascular risks. Therefore, it is important to be cautious about cardiovascular complications in patients receiving drugs that target PPAR-γ.

This review focused on studies that report the beneficial effects of pioglitazone on cognitive impairment and the associated mechanisms, such as changes in the expression or activity of proteins, mitochondrial function, neuroinflammation, oxidative stress, synaptic transmission, and thyroid function (Figure 1) (Table 2). We hope that our review will guide researchers to investigate these mechanisms in depth in the future and shed light on the potential clinical applications of pioglitazone for the treatment of cognitive disorders.

Figure 1 Mechanisms of pioglitazone in the improvement of memory impairment. The diagram depicts the factors that cause memory impairment (red lines) and the potential protective effects of pioglitazone (black lines).

Pioglitazone and Neuroinflammation

Neuroinflammation plays a key role in the pathogenesis of brain diseases12 and has been implicated in the development of neurodegenerative diseases and cognitive decline.13 In the central nervous system, inflammatory effectors derived from immune systems, as well as glial cells, specifically, microglia, act as sensors of disturbed brain tissue homeostasis and accumulate locally in response to neuronal cell injury.14 The production of inflammatory and neurotoxic mediators, such as cytokines, has been commonly linked to intracellular mechanisms, such as protein degradation, mitochondrial dysfunction, axonal transport defects, and apoptosis, which contribute to the progression of neurodegenerative diseases.12

The activation of PPARγ has been demonstrated to reduce neuroinflammation.15,16 PPARγ upregulates cluster of differentiation 36 or CD36, which is involved in the modulation of microglial activation and phenotypic differentiation, and this promotes the phagocytosis of apoptotic cells and contributes to the resolution of inflammation.15,17 Moreover, PPARγ mediates the downregulation of proinflammatory genes and reduces the production of proinflammatory chemokines, cytokines, and interleukins.15,18,19 Accordingly, PPARγ agonists have been found to exert anti-inflammatory effects by inducing an increase in the expression of anti-inflammatory-related genes in activated microglia and macrophages.20 In particular, the beneficial effects of the PPARγ agonist pioglitazone in the reduction of brain inflammatory responses have been detected in different in vivo and in vitro models of neurological conditions, such as AD, PD, Huntington’s disease, schizophrenia, and autism spectrum disorders.21–23 With regard to the mechanisms of pioglitazone, through its effect on PPARγ, it has been found to induce an antioxidant effect and reduce the levels of inflammatory molecules by inhibiting common inflammatory signaling pathways. For instance, pioglitazone reduced the inhibition of both nuclear factor kinase B (NF-kB) and p38 mitogen-activated protein kinase activity,24,25 which are known to ameliorate the neuroinflammatory response and oxidative stress.26,27

Apart from neuroinflammation, increased amyloid-β (Aβ) levels, oxidative stress, and mitochondrial dysfunction are other mechanisms associated with cognitive impairment in neurodegenerative diseases such as AD.21,28–30 The combination of pioglitazone with fenofibrate was found to be effective in ameliorating the behavioral, neurochemical, and histopathological changes associated with amyloidogenesis induced by increased Aβ levels in model AD mice, and the data indicate that this is a promising therapeutic approach in the management of AD complicated by diabetes and hypercholesterolemia.29 In another study, administration of pioglitazone was found to provide protection from lipopolysaccharide (LPS)-induced neuroinflammation and amyloidogenesis via targeting the glutamatergic and inflammatory pathways.30 Based on similar reports by several studies, it is speculated that pioglitazone causes a reduction in Aβ levels by decreasing Aβ formation or increasing its clearance.26,27,31

The literature so far indicates that downregulation of the levels of PPARγ and activation of microglia, brain inflammation, and amyloidogenesis are involved in the onset of cognitive deterioration in PD.32,33 Pioglitazone has an ameliorative effect on PD by decreasing microglial activation, downregulating PPARγ phosphorylation, increasing PPARγ expression, inhibiting beta-secretase 1 expression, and inhibiting Aβ production.34 There is considerable literature on the mechanisms of pioglitazone in improving cognitive dysfunction associated with PD, and discussing all of these is beyond the scope of the current review. Nonetheless, these mechanisms of cognitive impairment and its improvement by pioglitazone are likely to be true for other cognitive diseases too. Therefore, these findings warrant careful consideration for the treatment of cognitive decline associated with neurological diseases.

Pioglitazone and Mitochondria

Mitochondrial respiration is the main source of energy in cells.35 Mitochondria also regulate Ca2+ signaling, the generation of reactive oxygen species, synaptic function and plasticity, and the arbitration of cell survival,36 and mitochondrial dysfunction contributes to ageing and neurodegenerative diseases.37,38 Mitochondria are also involved in the long-term potentiation of synaptic transmission, a significant process in memory and learning.39–41 Cognitive impairment has been associated with increased brain mitochondrial oxidative stress, cellular energy depletion, impairment of mitochondrial electron transport and systems, and decreased mitochondrial integrity.42–44

Alterations in mitochondrial Ca2+ efflux, Aβ plaque accumulation, and intracellular deposition of hyperphosphorylated tau are known to accelerate memory deficits in AD.45 In addition, reduced levels of peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1-α), along with decreased ATP levels in the AD brain, trigger a decrease in mitochondrial density in different brain regions, but earlier data contradict this finding.46 Further, the level of PPARγ protein was found to be increased in the temporal cortex of AD patients.47,48 Therefore, this line of research requires further research in the future.

The activation of PPARγ stimulates the PGC1 signaling pathway, thus increasing mitochondrial function and mass and counteracting cognitive dysfunction.49 Through its agonistic effect on PPARγ, pioglitazone has been shown to improve mitochondrial bioenergetics and maintain mitochondrial function, thus promoting cognitive recovery.50,51 In particular, pioglitazone improves learning and memory by improving synaptic activity and reducing amyloid and tau pathologies.23 With regard to the underlying molecular mechanisms, pioglitazone was found to increase the expression of PGC-1α and the production of uncoupling protein 2, a mitochondrial protein that reduces reactive oxygen species, as well as COX-1, and prevents tumor necrosis factor-α (TNFα)-mediated effects.21,52

According to small-scale pilot studies on type 2 diabetes mellitus (DM2), pioglitazone increases cerebral blood flow and delays the onset of dementia; the findings indicate that its effects on cognition interacted with its insulin-lowering effects, even in cases without DM2.23 Several clinical trials conducted along this line have indicated that pioglitazone administration improved cognitive function.26 Further, combining pioglitazone with other antidiabetic drugs, as well as lowering its dose, had beneficial effects in terms of enhancing memory parameters.41,42 However, a recent study conducted on randomly selected participants of all age groups suggested that pioglitazone did not slow down the memory impairment and, in fact, was associated with deaths in test populations.23,53 This study indicated that cardiovascular risks, such as weight gain and edema, which increased the load on the system, were the major causes of mortality.23 Further, the lack of any effect of pioglitazone on dementia parameters was attributed to its unavailability in the brain cells due to its inability to cross the blood–brain barrier.15,23,53

MitoNEET is a protein in the mitochondrial outer membrane that plays a central role in the regulation of mitochondrial function and metabolism.54 Overexpression of mitoNEET has been found to lead to a significant reduction in inflammation and provide protection against oxidative stress.55 A recent study demonstrated that pioglitazone exerts neuroprotective and functional benefits following traumatic brain injury by targeting mitoNEET.56 Further, delayed pioglitazone treatment was found to be more effective in improving mitochondrial bioenergetics and attenuating brain atrophy, thus improving cognitive and motor performance.55 These findings could mean that the ameliorative effect of pioglitazone on cognitive impairment might involve its effect on mitoNEET. Therefore, it would be interesting to explore the mitoNEET-targeting mechanisms of pioglitazone in the treatment of cognitive disorders.

Role of Pioglitazone in Neurogenesis

Adult neurogenesis is defined as the formation of mature functional neurons from neural stem cells in the brain.57 It involves several events that begin with the division of a precursor cell and end with the presence of new mature functional neurons.57,58 Triggers, such as stress,59 bacterial LPS,60 and ischemia,17 cause neuroinflammatory processes in different brain regions such as the hippocampus and cortex.61 Microglia are immune cells located within the central nervous system that play a crucial role in the neuroinflammatory response.62 The activation of microglia and neuroinflammation can seriously affect the regeneration of neurons in different brain regions.63 Microglial activation caused by LPS administration or elevated levels of proinflammatory cytokines, such as interleukin-6 (IL-6) and TNFα, can disrupt hippocampal neurogenesis and thus affect cognitive and olfactory function, contributing to many neurodegenerative diseases.64

PPARγ is involved in the proliferation and differentiation of neural stem cells, controls the production of inflammatory mediators, and is essential in the regulation of brain development and repair following injury. PPARγ exists in most cell types, neurons, vessels, and astrocytes, where it performs multiple functions.65 In one study performed on experimental mice subjected to diesel exhaust, which causes microglial activation and neuroinflammation, it was found that diesel exhaust reduced adult hippocampal neurogenesis, with male mice showing fewer new neurons in the hippocampal subgranular zone, the olfactory bulb, and the subventricular zone, and female mice showing fewer new neurons in the olfactory bulb.66 However, administration of the PPARα agonist pioglitazone suppressed the effects of diesel exhaust on microglia and neuroinflammation.67 In another study, rats were administered LPS, which is known to cause inflammation by promoting the synthesis of the cytokines IL-1, IL-6, and TNF-α, leading to cognitive impairment in rats.64,68 Administration of pioglitazone prior to LPS exposure protected rats from LPS-induced learning and memory impairment through the regulation of secreted cytokines and the improvement of oxidative stress and brain-derived neurotrophic factor (BDNF) levels.69 However, despite the well-documented anti-inflammatory effects of pioglitazone, it can cause cell death when administered at high concentrations.70 Therefore, its application in the treatment of PD, AD, DM2, and other diseases associated with cognitive impairment should be assessed with caution.

Effect of Pioglitazone on Protein Kinases

Protein kinases and phosphatases are phosphotransferase enzymes that catalyze the transfer of phosphates between substrates. A protein kinase catalyzes the transfer of γ-phosphate from ATP (or GTP) to its protein substrates, thus regulating the reactions that modulate the structures and functions of many cellular proteins within cells.71 An imbalance between kinase and/or phosphatase functions has been considered to be the primary cause of deregulated protein phosphorylation. AD is a progressive neurodegenerative disorder characterized by severe impairment of memory and cognitive function72 that results from the accumulation of misfolded Aβ and phosphorylated tau proteins in the aging brain.73 These misfolded proteins form intraneuronal neurofibrillary tangles74 that lead to oxidative and inflammatory damage, resulting in energy failure and synaptic dysfunction.75

In the AD brain, the expression or activity of kinases, such as Akt, extracellular signal-regulated kinase 1 and 2 (ERK1/2), cAMP-dependent protein kinase (PKA), glycogen synthase kinase-3β (GSK-3β), p70S6 kinase, and cyclin-dependent protein kinases 5 (Cdk5), was found to be increased, whereas that of several protein phosphatases, such as PP1, PP2A, and PP5, was found to be decreased.76,77 One study demonstrated that pioglitazone could inhibit the in vitro phosphorylation of PPARγ by inhibiting the expression of cyclin-dependent kinase 5 (CDK5), which increases the expression of the insulin-degrading enzyme and inhibits β-amyloid cleavage enzyme 1 through PPARγ target genes, leading to Aβ degradation and reduced Aβ production.78 In accordance with these findings, reduction of Aβ levels in the brain exerted neuroprotective effects in an AD model.33 Importantly, the use of pioglitazone improved spatial learning, enhanced AKT signaling, and attenuated tau hyperphosphorylation and neuroinflammation79 through enhanced microglial uptake of Aβ.80,81

PD is a neurodegenerative disorder characterized by non-motor and motor symptoms caused by a depletion of striatal dopamine owing to the degradation of dopaminergic neurons in the substantia nigra.82,83 The available data from several previous studies strongly implicate the dysfunction of kinase activities and phosphorylation pathways in the pathogenesis of PD.6,32 Some of the important protein kinases associated with increased risk of PD are PTEN (phosphatase and tensin homolog)-induced putative kinase 1 (PINK1), and leucine-rich repeat kinase 2 (LRRK2).29,32 In particular, PINK1 and LRRK2—along with their associated protein kinase B (AKT) and c-Jun N-terminal kinase (JNK) signaling pathways—are being intensively studied with respect to their relationship to PD.31,77

Some studies have demonstrated that treatment with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine activates caspase-3, leading to neuronal death by apoptosis; however, treatment with neuroprotective drugs such as pioglitazone inhibits the activity of caspase-3 and reduces neuronal damage.84,85 Previous studies have also indicated that pioglitazone protects dopaminergic neurons by inhibiting abnormal microglia activation, interfering with the phosphorylation of JNK and NF-κB, and suppressing cyclooxygenase 2 expression and subsequent prostaglandin E(2) synthesis.86

Thus, the beneficial effects of pioglitazone in PD and AD seem to be linked to its effects on the regulation of protein kinases, and therefore, its protein kinase-related mechanisms should be explored further in the treatment of cognitive disorders.

Pioglitazone and Oxidative Stress

Chronic exposure to stress increases the levels of stress hormones, such as cortisol, and leads to several health complications, including alterations in brain functions.87,88 In addition, severe and long-term elevation in the levels of stress hormones can induce changes in normal brain structure and function,89 ultimately affecting cognitive function and leading to memory impairment.90,91 Several studies conducted in experimental rats have revealed that stress alters the expression and function of proteins, such as BDNF and CaMKII, which affect cognitive function.92–94 Earlier studies indicated that activation of the NMDA receptor in glutamatergic excitatory pathways following a behavioral experience is essential for long-term memory (LTM) storage.95 When this receptor is activated, calcium enters into the neurons and binds to calmodulin to form the Ca2C/CaM complex, which is recognized by multiple enzymes and induces a molecular signaling cascade whose main function is to reshape synaptic structure and physiology, as well as regulate the expression of genes necessary for the formation of LTM.100 It is considered that CaMKII is one of the main targets of Ca2C/CaM, and accordingly, CaMKII activity is increased in response to learning and its inhibition causes LTM impairment.

Interestingly, pioglitazone has been reported to exert antioxidant activity,96 and treatment with pioglitazone was found to rescue the cardiovascular system and cognitive impairment caused by elevated levels of oxidative stress.97 The underlying mechanisms of the cardioprotective effects and improvement in memory impairment induced by pioglitazone were found to be mediated via modulation of the antioxidant protein TRX198 and increased expression of BDNF and CaMKII. In particular, CaMKII binding and pioglitazone-induced phosphorylation has been found to regulate autonomous activity and the location and/or transport of proteins in the post-synaptic density, including the NMDA receptor, synapsin 1, F-actin, and calcium channels, to regions of interest.99 In addition, the expression of p47phox and gp91phox, which is increased under conditions of oxidative stress, was reduced following treatment with pioglitazone.96 Further, several studies have pointed to the effect of pioglitazone on increased expression of the antioxidant protein PON2.100,101

Malondialdehyde (MDA) is one of the products generated from the peroxidation of fatty acids, and an increase in the levels of reactive oxygen species has been positively correlated with overexpression of MDA.102,103 Pioglitazone has also been found to reduce oxidative stress through a reduction in the levels of MDA and inhibition of ROS production and inflammatory pathways; this further verifies its potent antioxidant activity.104 All these findings indicate the role of pioglitazone against stress in the brain via regulation of TRX1, p47phox, and gp91phox activity, and the levels of MDA and ROS.

Pioglitazone and Thyroid Function

Thyroid hormones, such as thyroxine (T4) and triiodothyronine (T3), regulate many physiological functions.105 These hormones play essential roles in brain development, particularly in neurogenesis and synaptogenesis.106,107 More specifically, T3 and T4 are produced by thyroid follicular cells in the thyroid gland and then released into the bloodstream, from where they are capable of crossing the blood–brain barrier108,109 and ultimately binding to thyroid hormone receptor (THRs). THRs are nuclear receptors that can be classified into two isoforms, THRα and THRβ,110 the distribution of which differs among tissues. The THRα1 receptor is predominantly expressed in the heart and skeletal muscle,111 whereas THRβ1 is primarily expressed in the liver, kidney, and brain.112 Of note, THRs are abundantly expressed in the hippocampus, the structure within which memory is formed in the brain.107 THRα-knockout mice exhibited memory impairment; this confirms the role of THRα in memory processes.113,114 Hence, in hyperthyroidism, hypothyroidism, and cretinism, which are characterized by abnormal levels of thyroid hormones,115 hippocampal functionality might be altered and potentially result in cognitive dysfunction.116 Indeed, neuroimaging studies have reported that in patients with hypothyroidism, the structure and function of the hippocampus were altered.117–119

Treatment with pioglitazone has been shown to enhance the activation of THRs and, thus, learning and memory processes.112 Memory impairment caused by hypothyroidism was improved by pioglitazone via a reduction in oxidative stress that was attributed to the increased expression of superoxide dismutase and catalase.120 In addition, pioglitazone treatment in patients with DM2 was associated with reduced levels of T4 and increased levels of TSH; this indicates the possible activation of THRs through the activation of PPARγ. Thus, one of the mechanisms by which pioglitazone improves memory impairment caused by diabetes and hypothyroidism may involve the activation of THRs. However, the details of these mechanisms underlying improvement in cognitive impairment by treatment with pioglitazone in patients with hypothyroidism require further investigation.

Pioglitazone and Synaptic Transmission

Synaptic transmission is a physiological process via which neurons communicate with other cells through synapses.121 This communication occurs via chemical and electrical synaptic transmission.122 Chemical synaptic transmission occurs when a neurotransmitter is released into the synapse and binds to specific postsynaptic receptors, whereas electrical synaptic transmission occurs when electrical signals transferred through neurons are involved in neurotransmitter release.122,123 In learning and memory processes, neurons are stimulated and lead to the firing of other neurons with action potential, thus enhancing the release of neurotransmitters through the activation of calcium channels in presynaptic neurons.124,125 Alterations in these processes, whether they occur in pre- or postsynaptic neurons, can impair memory function.126 For instance, changes in the expression and function of proteins have been observed in both pre- and postsynaptic neurons in AD and PD.127

Synaptic transmission in ischemic neurons is impaired as a result of the reduction in the activity of PI3K/Akt and Nrf2/ARE pathways, and pioglitazone treatment has been found to increase the pro-survival PI3K/Akt pathway by increasing the phosphorylation of Akt in Ser473 and GSK-3β in Ser9 residues. This further improved the activity of the Nrf2/ARE antioxidant pathway.128 It has also been shown that lipoprotein receptor-related protein 1 (LRP1) and nuclear factor kappa B (NF-κB) p65 were downregulated in AD, but they were upregulated by pioglitazone treatment.6,129 All together, these findings demonstrate that pioglitazone can improve synaptic transmission by increasing the expression and phosphorylation of LRP1, NF-κB p65, Akt in Ser473, and GSK-3β in Ser9 residues, and increasing the activity of the Nrf2/ARE pathways. Thus, the mechanisms via which pioglitazone improves memory impairment in AD and PD, and perhaps metabolic diseases such as DM2, might involve its effects on the LRP1, NF-κB p65, Akt, GSK-3β, and Nrf2/ARE pathways, so these pathways warrant further research in the future with regard to the identification of potential treatment targets.

Conclusion

The findings reported in the studies included in the current review demonstrate that pioglitazone can improve memory function via the activation of PPAR receptors. The included studies have indicated that PPAR receptors play a role in ameliorating inflammation, oxidative stress, amyloidogenesis, and hypothyroidism, and enhancing neurogenesis, synaptic plasticity, and mitochondrial function; further, the effects of pioglitazone on these processes are also evident. Based on these findings, we believe that further in-depth investigations into these mechanisms will help establish the potential of pioglitazone in terms of its clinical applicability in the management of cognitive disorders (Figure 2).

Figure 2 Mechanism of protective effect of pioglitazone on memory impairment.

Disclosure

The authors report no conflicts of interest related to this work.

References

1. Hong F, Pan S, Guo Y, Xu P, Zhai Y. PPARs as nuclear receptors for nutrient and energy metabolism. Molecules. 2019;24:14. doi:10.3390/molecules24142545

2. Villacorta L, Schopfer Francisco J, Zhang J, Freeman Bruce A, Chen YE. PPARγ and its ligands: therapeutic implications in cardiovascular disease. Clin Sci. 2009;116(3):205–218. doi:10.1042/CS20080195

3. Chang KL, Pee HN, Yang S, Ho PC. Influence of drug transporters and stereoselectivity on the brain penetration of pioglitazone as a potential medicine against Alzheimer's’s disease. Sci Rep. 2015;5(1). doi:10.1038/srep09000

4. Takechi R, Lam V, Brook E, et al. Blood-brain barrier dysfunction precedes cognitive decline and neurodegeneration in diabetic insulin resistant mouse model: an implication for causal link. Front Aging Neurosci. 2017;9. doi:10.3389/fnagi.2017.00399

5. Liu M, Bachstetter AD, Cass WA, Lifshitz J, Bing G. Pioglitazone attenuates neuroinflammation and promotes dopaminergic neuronal survival in the nigrostriatal system of rats after diffuse brain injury. J Neurotrauma. 2017;34(2):414–422. doi:10.1089/neu.2015.4361

6. Seok H, Lee M, Shin E, et al. Low-dose pioglitazone can ameliorate learning and memory impairment in a mouse model of dementia by increasing LRP1 expression in the hippocampus. Sci Rep. 2019;9(1). doi:10.1038/s41598-019-40736-x.

7. Fernandez-Martos CM, Atkinson RAK, Chuah MI, King AE, Vickers JC. Combination treatment with leptin and pioglitazone in a mouse model of Alzheimer's’s disease. Alzheimer's’s Dementia. 2017;3(1):92–106.

8. Hanyu H, Sato T, Kiuchi A, Sakurai H, Iwamoto T. Pioglitazone improved cognition in a pilot study on patients with Alzheimer’s disease and mild cognitive impairment with diabetes mellitus. J Am Geriatr Soc. 2009;57(1):177–179. doi:10.1111/j.1532-5415.2009.02067.x

9. Hildreth KL, Van Pelt RE, Moreau KL, et al. Effects of pioglitazone or exercise in older adults with mild cognitive impairment and insulin resistance: a pilot study. Dement Geriatr Cogn Dis Extra. 2015;5(1):51–63. doi:10.1159/000371509

10. Kline AE, Yin -Q-Q, Pei -J-J, et al. Pioglitazone improves cognitive function via increasing insulin sensitivity and strengthening antioxidant defense system in fructose-drinking insulin resistance rats. PLoS One. 2013;8(3):548.

11. Wang Y, Zhao W, Li G, et al. Neuroprotective Effect and Mechanism of Thiazolidinedione on Dopaminergic Neurons In Vivo and In Vitro in Parkinson’s Disease. PPAR Res. 2017;2017:1–12. doi:10.1155/2017/6561701

12. Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflammation: the role and consequences. Neurosci Res. 2014;79:1–12. doi:10.1016/j.neures.2013.10.004

13. Gorelick PB. Role of inflammation in cognitive impairment: results of observational epidemiological studies and clinical trials. Ann N Y Acad Sci. 2010;1207(1):155–162. doi:10.1111/j.1749-6632.2010.05726.x

14. McManus RM, Heneka MT. Role of neuroinflammation in neurodegeneration: new insights. Alzheimer's’s Res Therapy. 2017;9(1):7.

15. Villapol S. Roles of Peroxisome Proliferator-Activated Receptor Gamma on Brain and Peripheral Inflammation. Cell Mol Neurobiol. 2017;38(1):121–132. doi:10.1007/s10571-017-0554-5

16. Zhang Z, Yuan H, Zhao H, Qi B, Li F, An L. PPARγ activation ameliorates postoperative cognitive decline probably through suppressing hippocampal neuroinflammation in aged mice. Int Immunopharmacol. 2017;43:53–61. doi:10.1016/j.intimp.2016.12.003

17. Ballesteros I, Cuartero MI, Pradillo JM, et al. Rosiglitazone-induced CD36 up-regulation resolves inflammation by PPAR and 5-LO-dependent pathways. J Leukoc Biol. 2013;95(4):587–598. doi:10.1189/jlb.0613326

18. Kapadia R. Mechanisms of anti-inflammatory and neuroprotective actions of PPAR-gamma agonists. Front Biosci. 2008;13(13):1813. doi:10.2741/2802

19. Farooqui AA, Horrocks LA, Farooqui T. Modulation of inflammation in brain: a matter of fat. J Neurochem. 2006;101(3):577–599. doi:10.1111/j.1471-4159.2006.04371.x

20. Bouhlel MA, Derudas B, Rigamonti E, et al. PPARγ Activation Primes Human Monocytes into Alternative M2 Macrophages with Anti-inflammatory Properties. Cell Metab. 2007;6(2):137–143. doi:10.1016/j.cmet.2007.06.010

21. d’Angelo M, Castelli V, Catanesi M, et al. PPARγ and Cognitive Performance. Int J Mol Sci. 2019;20(20):5068. doi:10.3390/ijms20205068

22. King E, O’Brien J, Donaghy P, et al. Inflammation in mild cognitive impairment due to Parkinson’s disease, Lewy body disease, and Alzheimer's’s disease. Int J Geriatr Psychiatry. 2019;34(8):1244–1250. doi:10.1002/gps.5124

23. Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's’s Disease. Front Neurosci. 2021;15. doi:10.3389/fnins.2021.666958

24. Ciaramella V, Sasso FC, Di Liello R, et al. Activity and molecular targets of pioglitazone via blockade of proliferation, invasiveness and bioenergetics in human NSCLC. J Exp Clin Cancer Res. 2019;38(1). doi:10.1186/s13046-019-1176-1.

25. Ojcius DM, Wang Y, Yu B, et al. Pioglitazone ameliorates glomerular NLRP3 inflammasome activation in apolipoprotein E knockout mice with diabetes mellitus. PLoS One. 2017;12(7):e0181248.

26. Dias V, Junn E, Mouradian MM. The Role of Oxidative Stress in Parkinson’s Disease. J Parkinson’s Dis. 2013;3(4):461–491. doi:10.3233/JPD-130230

27. Wei Z, Li X, Li X, Liu Q, Cheng Y. Oxidative Stress in Parkinson’s Disease: a Systematic Review and Meta-Analysis. Front Mol Neurosci. 2018;11. doi:10.3389/fnmol.2018.00236

28. Castelli V, Benedetti E, Antonosante A, et al. Neuronal Cells Rearrangement During Aging and Neurodegenerative Disease: metabolism, Oxidative Stress and Organelles Dynamic. Front Mol Neurosci. 2019;12. doi:10.3389/fnmol.2019.00132

29. Passamonti L, Tsvetanov KA, Jones PS, et al. Neuroinflammation and Functional Connectivity in Alzheimer's’s Disease: interactive Influences on Cognitive Performance. J Neurosci. 2019;39(36):7218–7226. doi:10.1523/JNEUROSCI.2574-18.2019

30. Mondragón-Rodríguez S, Perry G, Zhu X, Moreira PI, Acevedo-Aquino MC, Williams S. Phosphorylation of Tau Protein as the Link between Oxidative Stress, Mitochondrial Dysfunction, and Connectivity Failure: implications for Alzheimer's’s Disease. Oxid Med Cell Longev. 2013;2013:1–6. doi:10.1155/2013/940603

31. Gad ES, Zaitone SA, Moustafa YM. Pioglitazone and exenatide enhance cognition and downregulate hippocampal beta amyloid oligomer and microglia expression in insulin-resistant rats. Can J Physiol Pharmacol. 2016;94(8):819–828. doi:10.1139/cjpp-2015-0242

32. Heneka MT, Sastre M, Dumitrescu-Ozimek L, et al. Acute treatment with the PPARγ agonist pioglitazone and ibuprofen reduces glial inflammation and Aβ1–42 levels in APPV717I transgenic mice. Brain. 2005;128(6):1442–1453. doi:10.1093/brain/awh452

33. Quan Q, Qian Y, Li X, Li M. Pioglitazone Reduces β Amyloid Levels via Inhibition of PPARγ Phosphorylation in a Neuronal Model of Alzheimer's’s Disease. Front Aging Neurosci. 2019;11. doi:10.3389/fnagi.2019.00178

34. Falcone R, Marilena Florio T, Giacomo ED, et al. PPARβ/δ and γ in a Rat Model of Parkinson’s Disease: possible Involvement in PD Symptoms. J Cell Biochem. 2015;116(5):844–855. doi:10.1002/jcb.25041

35. Rangaraju V, Calloway N, Ryan Timothy A. Activity-Driven Local ATP Synthesis Is Required for Synaptic Function. Cell. 2014;156(4):825–835. doi:10.1016/j.cell.2013.12.042

36. Mattson MP, Gleichmann M, Cheng A. Mitochondria in Neuroplasticity and Neurological Disorders. Neuron. 2008;60(5):748–766. doi:10.1016/j.neuron.2008.10.010

37. Monzio Compagnoni G, Di Fonzo A, Corti S, Comi GP, Bresolin N, Masliah E. The Role of Mitochondria in Neurodegenerative Diseases: the Lesson from Alzheimer's’s Disease and Parkinson’s Disease. Mol Neurobiol. 2020;57(7):2959–2980. doi:10.1007/s12035-020-01926-1

38. Wang Y, Xu E, Musich PR, Lin F. Mitochondrial dysfunction in neurodegenerative diseases and the potential countermeasure. CNS Neurosci Ther. 2019;25(7):816–824. doi:10.1111/cns.13116

39. Williams JM, Thompson VL, Mason-Parker SE, Abraham WC, Tate WP. Synaptic activity-dependent modulation of mitochondrial gene expression in the rat hippocampus. Mol Brain Res. 1998;60(1):50–56. doi:10.1016/S0169-328X(98)00165-X

40. Calabresi P, Ammassari-Teule M, Gubellini P, et al. A synaptic mechanism underlying the behavioral abnormalities induced by manganese intoxication. Neurobiol Dis. 2001;8(3):419–432. doi:10.1006/nbdi.2000.0379

41. Li Z, Jo J, Jia J-M, et al. Caspase-3 Activation via Mitochondria Is Required for Long-Term Depression and AMPA Receptor Internalization. Cell. 2010;141(5):859–871. doi:10.1016/j.cell.2010.03.053

42. Pintana H, Apaijai N, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Effects of metformin on learning and memory behaviors and brain mitochondrial functions in high fat diet induced insulin resistant rats. Life Sci. 2012;91(11–12):409–414. doi:10.1016/j.lfs.2012.08.017

43. Boscolo A, Starr JA, Sanchez V, et al. The abolishment of anesthesia-induced cognitive impairment by timely protection of mitochondria in the developing rat brain: the importance of free oxygen radicals and mitochondrial integrity. Neurobiol Dis. 2012;45(3):1031–1041. doi:10.1016/j.nbd.2011.12.022

44. Apaijai N, Sriwichaiin S, Phrommintikul A, et al. Cognitive impairment is associated with mitochondrial dysfunction in peripheral blood mononuclear cells of elderly population. Sci Rep. 2020;10(1). doi:10.1038/s41598-020-78551-4.

45. Jadiya P, Kolmetzky DW, Tomar D, et al. Impaired mitochondrial calcium efflux contributes to disease progression in models of Alzheimer's’s disease. Nat Commun. 2019;10:1. doi:10.1038/s41467-019-11813-6

46. Wareski P, Vaarmann A, Choubey V, et al. PGC-1α and PGC-1Β regulate mitochondrial density in neurons. J Biol Chem. 2009;284(32):21379–21385. doi:10.1074/jbc.M109.018911

47. Kim EJ, Kwon KJ, Park J-Y, Lee SH, Moon C-H, Baik EJ. Effects of peroxisome proliferator-activated receptor agonists on LPS-induced neuronal death in mixed cortical neurons: associated with iNOS and COX-2. Brain Res. 2002;941(1–2):1–10. doi:10.1016/S0006-8993(02)02480-0

48. Luna-Medina R, Cortes-Canteli M, Alonso M, Santos A, Martínez A, Perez-Castillo A. Regulation of inflammatory response in neural cells in vitro by thiadiazolidinones derivatives through peroxisome proliferator-activated receptor γ activation. J Biol Chem. 2005;280(22):21453–21462. doi:10.1074/jbc.M414390200

49. Chiang M-C, Cheng Y-C, Chen H-M, Liang Y-J, Yen C-H. Rosiglitazone promotes neurite outgrowth and mitochondrial function in N2A cells via PPARgamma pathway. Mitochondrion. 2014;14:7–17. doi:10.1016/j.mito.2013.12.003

50. Yonutas HM, Hubbard WB, Pandya JD, Vekaria HJ, Geldenhuys WJ, Sullivan PG. Bioenergetic restoration and neuroprotection after therapeutic targeting of mitoNEET: new mechanism of pioglitazone following traumatic brain injury. Exp Neurol. 2020;327:113243.

51. Sauerbeck A, Gao J, Readnower R, et al. Pioglitazone attenuates mitochondrial dysfunction, cognitive impairment, cortical tissue loss, and inflammation following traumatic brain injury. Exp Neurol. 2011;227(1):128–135. doi:10.1016/j.expneurol.2010.10.003

52. De Nuccio C, Bernardo A, Cruciani C, De Simone R, Visentin S, Minghetti L. Peroxisome proliferator activated receptor-γ agonists protect oligodendrocyte progenitors against tumor necrosis factor-alpha-induced damage: effects on mitochondrial functions and differentiation. Exp Neurol. 2015;271:506–514. doi:10.1016/j.expneurol.2015.07.014

53. Nicolakakis N. The nuclear receptor PPARγ as a therapeutic target for cerebrovascular and brain dysfunction in Alzheimer's’s disease. Front Aging Neurosci. 2010;2. doi:10.3389/fnagi.2010.00021

54. Wang Y, Landry AP, Ding H. The mitochondrial outer membrane protein mitoNEET is a redox enzyme catalyzing electron transfer from FMNH2 to oxygen or ubiquinone. J Biol Chem. 2017;292(24):10061–10067. doi:10.1074/jbc.M117.789800

55. Hubbard WB, Spry ML, Gooch JL, et al. Clinically relevant mitochondrial-targeted therapy improves chronic outcomes after traumatic brain injury. Brain. 2021;144(12):3788–3807. doi:10.1093/brain/awab341

56. Rabchevsky A, Patel S, Sullivan P. Targeting mitoNEET with pioglitazone for therapeutic neuroprotection after spinal cord injury. Neural Regeneration Res. 2017;12(11):1807. doi:10.4103/1673-5374.219040

57. Ming G-l SH. Adult Neurogenesis in the Mammalian Brain: significant Answers and Significant Questions. Neuron. 2011;70(4):687–702. doi:10.1016/j.neuron.2011.05.001

58. Kempermann G, Song H, Gage FH. Neurogenesis in the Adult Hippocampus. Cold Spring Harb Perspect Biol. 2015;7(9):a018812. doi:10.1101/cshperspect.a018812

59. Han A, Yeo H, Park M-J, et al. IL-4/10 prevents stress vulnerability following imipramine discontinuation. J Neuroinflammation. 2015;12(1). doi:10.1186/s12974-015-0416-3.

60. Swiergiel AH, Dunn AJ. Effects of interleukin-1β and lipopolysaccharide on behavior of mice in the elevated plus-maze and open field tests. Pharmacol Biochem Behav. 2007;86(4):651–659. doi:10.1016/j.pbb.2007.02.010

61. Qin L, Wu X, Block ML, et al. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007;55(5):453–462. doi:10.1002/glia.20467

62. Hanisch U-K KH. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10(11):1387–1394. doi:10.1038/nn1997

63. Carpentier PA, Palmer TD. Immune Influence on Adult Neural Stem Cell Regulation and Function. Neuron. 2009;64(1):79–92. doi:10.1016/j.neuron.2009.08.038

64. Mani V, Arfeen M, Ali HM, A-MH A-M, Aldubayan M, Alhowail A. Neuroprotective Effect of Clobenpropit against Lipopolysaccharide-Induced Cognitive Deficits via Attenuating Neuroinflammation and Enhancing Mitochondrial Functions in Mice. Brain Sci. 2021;11(12):1617. doi:10.3390/brainsci11121617

65. Heming M, Gran S, Jauch S-L, et al. Peroxisome Proliferator-Activated Receptor-γ Modulates the Response of Macrophages to Lipopolysaccharide and Glucocorticoids. Front Immunol. 2018;9. doi:10.3389/fimmu.2018.00893

66. Sui Y, Vermeulen R, Hökfelt T, Horne MK, Stanić D. Female mice lacking cholecystokinin 1 receptors have compromised neurogenesis, and fewer dopaminergic cells in the olfactory bulb. Front Cell Neurosci. 2013;7. doi:10.3389/fncel.2013.00013

67. Zhao Q, Wu X, Yan S, et al. The antidepressant-like effects of pioglitazone in a chronic mild stress mouse model are associated with PPARγ-mediated alteration of microglial activation phenotypes. J Neuroinflammation. 2016;13(1). doi:10.1186/s12974-016-0728-y.

68. Ng A, Tam WW, Zhang MW, et al. IL-1β, IL-6, TNF- α and CRP in Elderly Patients with Depression or Alzheimer's’s disease: systematic Review and Meta-Analysis. Sci Rep. 2018;8(1). doi:10.1038/s41598-018-30487-6.

69. Giannini S, Serio M, Galli A. Pleiotropic effects of thiazolidinediones: taking a look beyond antidiabetic activity. J Endocrinol Invest. 2014;27(10):982–991. doi:10.1007/BF03347546

70. Wada K, Nakajima A, Katayama K, et al. Peroxisome Proliferator-activated Receptor γ-mediated Regulation of Neural Stem Cell Proliferation and Differentiation. J Biol Chem. 2006;281(18):12673–12681. doi:10.1074/jbc.M513786200

71. Ubersax JA, Ferrell Jr JJE. Mechanisms of specificity in protein phosphorylation. Nat Rev Mol Cell Biol. 2007;8(7):530–541. doi:10.1038/nrm2203

72. Tarawneh R, Holtzman DM. The Clinical Problem of Symptomatic Alzheimer's Disease and Mild Cognitive Impairment. Cold Spring Harb Perspect Med. 2012;2(5):a006148–a006148. doi:10.1101/cshperspect.a006148

73. Rao RV, Bredesen DE. Misfolded proteins, endoplasmic reticulum stress and neurodegeneration. Curr Opin Cell Biol. 2004;16(6):653–662. doi:10.1016/j.ceb.2004.09.012

74. Takahashi RH, Nagao T, Gouras GK. Plaque formation and the intraneuronal accumulation of β-amyloid in Alzheimer's’s disease. Pathol Int. 2017;67(4):185–193. doi:10.1111/pin.12520

75. Barage SH, Sonawane KD. Amyloid cascade hypothesis: pathogenesis and therapeutic strategies in Alzheimer's’s disease. Neuropeptides. 2015;52:1–18. doi:10.1016/j.npep.2015.06.008

76. Iqbal K, Alzheimer's Review G-I-I. Series: Alzheimer neurofibrillary degeneration: significance, etiopathogenesis, therapeutics and prevention. J Cell Mol Med. 2007;12(1):38–55. doi:10.1111/j.1582-4934.2008.00225.x

77. Perluigi M, Barone E, Di Domenico F, Butterfield DA. Aberrant protein phosphorylation in Alzheimer's disease brain disturbs pro-survival and cell death pathways. Biochimica et Biophysica Acta. 2016;1862(10):1871–1882. doi:10.1016/j.bbadis.2016.07.005

78. Quan Q, Li X, Feng J, Hou J, Li M, Zhang B. Ginsenoside Rg1 reduces β‑amyloid levels by inhibiting CD5K‑induced PPARγ phosphorylation in a neuron model of Alzheimer's’s disease. Mol Med Rep. 2020. doi:10.3892/mmr.2020.11424

79. Yu Y, Li X, Blanchard J, et al. Insulin sensitizers improve learning and attenuate tau hyperphosphorylation and neuroinflammation in 3xTg-AD mice. J Neural Transm. 2014;122(4):593–606. doi:10.1007/s00702-014-1294-z

80. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPAR /RXR -Induced and CD36-Mediated Microglial Amyloid- Phagocytosis Results in Cognitive Improvement in Amyloid Precursor Protein/Presenilin 1 Mice. J Neurosci. 2012;32(48):17321–17331. doi:10.1523/JNEUROSCI.1569-12.2012

81. Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms Underlying the Rapid Peroxisome Proliferator-Activated Receptor- -Mediated Amyloid Clearance and Reversal of Cognitive Deficits in a Murine Model of Alzheimer's’s Disease. J Neurosci. 2012;32(30):10117–10128. doi:10.1523/JNEUROSCI.5268-11.2012

82. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443(7113):787–795. doi:10.1038/nature05292

83. Navarro A. Brain mitochondrial dysfunction in aging, neurodegeneration and Parkinson’s disease. Front Aging Neurosci. 2010. doi:10.3389/fnagi.2010.00034

84. Hughes RE, Nikolic K, Ramsay RR. One for All? Hitting Multiple Alzheimer's’s Disease Targets with One Drug. Front Neurosci. 2016;10. doi:10.3389/fnins.2016.00177

85. Walker DG, Lue L-F, Serrano G, et al. Altered Expression Patterns of Inflammation-Associated and Trophic Molecules in Substantia Nigra and Striatum Brain Samples from Parkinson’s Disease, Incidental Lewy Body Disease and Normal Control Cases. Front Neurosci. 2016;9. doi:10.3389/fnins.2015.00507

86. Lanzillotta A, Porrini V, Bellucci A, et al. NF-κB in Innate Neuroprotection and Age-Related Neurodegenerative Diseases. Front Neurol. 2015;6. doi:10.3389/fneur.2015.00098

87. McEwen BS. Central effects of stress hormones in health and disease: understanding the protective and damaging effects of stress and stress mediators. Eur J Pharmacol. 2008;583(2–3):174–185. doi:10.1016/j.ejphar.2007.11.071

88. Felemban SG, Aldubayan MA, Alhowail AH, Almami IS. Vitamin B17 Ameliorates Methotrexate-Induced Reproductive Toxicity, Oxidative Stress, and Testicular Injury in Male Rats. Oxid Med Cell Longev. 2020;2020:4372719. doi:10.1155/2020/4372719

89. JNd S-T, Marin M-F, Sindi S, Lupien SJ. Effects of stress hormones on the brain and cognition: evidence from normal to pathological aging. Dementia Neuropsychol. 2011;5(1):8–16. doi:10.1590/S1980-57642011DN05010003

90. Ouanes S, High PJ. Cortisol and the Risk of Dementia and Alzheimer's’s Disease: a Review of the Literature. Front Aging Neurosci. 2019;11. doi:10.3389/fnagi.2019.00043

91. Peavy GM, Salmon DP, Jacobson MW, et al. Effects of Chronic Stress on Memory Decline in Cognitively Normal and Mildly Impaired Older Adults. Am J Psychiatry. 2009;166(12):1384–1391. doi:10.1176/appi.ajp.2009.09040461

92. Zhang L, Zhang H-Q, Liang X-Y, Zhang H-F, Zhang T, Liu F-E. Melatonin ameliorates cognitive impairment induced by sleep deprivation in rats: role of oxidative stress, BDNF and CaMKII. Behav Brain Res. 2013;256:72–81. doi:10.1016/j.bbr.2013.07.051

93. Sałaciak K, Koszałka A, Żmudzka E, Pytka K. The Calcium/Calmodulin-Dependent Kinases II and IV as Therapeutic Targets in Neurodegenerative and Neuropsychiatric Disorders. Int J Mol Sci. 2021;22(9):4307. doi:10.3390/ijms22094307

94. Alzoubi KH, Srivareerat M, Aleisa AM, Alkadhi KA. Chronic Caffeine Treatment Prevents Stress-Induced LTP Impairment: the Critical Role of Phosphorylated CaMKII and BDNF. J Mol Neurosci. 2012;49(1):11–20. doi:10.1007/s12031-012-9836-z

95. Zalcman G, Federman N, Romano A. CaMKII Isoforms in Learning and Memory: localization and Function. Front Mol Neurosci. 2018;11. doi:10.3389/fnmol.2018.00445

96. Dobrian AD, Schriver SD, Khraibi AA, Prewitt RL. Pioglitazone Prevents Hypertension and Reduces Oxidative Stress in Diet-Induced Obesity. Hypertension. 2004;43(1):48–56. doi:10.1161/01.HYP.0000103629.01745.59

97. Govindarajulu M, Pinky PD, Bloemer J, Ghanei N, Suppiramaniam V, Amin R. Signaling Mechanisms of Selective PPARγ Modulators in Alzheimer's’s Disease. PPAR Res. 2018;2018:1–20. doi:10.1155/2018/2010675

98. Bilginoglu A. Cardiovascular protective effect of pioglitazone on oxidative stress in rats with metabolic syndrome. J Chine Med Assoc. 2019;82(6):452–456. doi:10.1097/JCMA.0000000000000103

99. Malik Zulfiqar A, Stein Ivar S, Navedo Manuel F, Hell Johannes W. Mission CaMKIIγ: shuttle Calmodulin from Membrane to Nucleus. Cell. 2014;159(2):235–237. doi:10.1016/j.cell.2014.09.023

100. Blackburn JK, Curry DW, Thomsen AN, Roth RH, Elsworth JD. Pioglitazone activates paraoxonase-2 in the brain: a novel neuroprotective mechanism. Exp Neurol. 2020;327:113234. doi:10.1016/j.expneurol.2020.113234

101. Blackburn JK, Jamwal S, Wang W, Elsworth JD. Pioglitazone transiently stimulates paraoxonase-2 expression in male nonhuman primate brain: implications for sex-specific therapeutics in neurodegenerative disorders. Neurochem Int. 2022;152:105222. doi:10.1016/j.neuint.2021.105222

102. Shields HJ, Traa A, Van Raamsdonk JM. Beneficial and Detrimental Effects of Reactive Oxygen Species on Lifespan: a Comprehensive Review of Comparative and Experimental Studies. Front Cell Dev Biol. 2021;9. doi:10.3389/fcell.2021.628157

103. Elgharabawy RM, Alhowail AH, Emara AM, Aldubayan MA, Ahmed AS. The impact of chicory (Cichorium intybus L.) on hemodynamic functions and oxidative stress in cardiac toxicity induced by lead oxide nanoparticles in male rats. Biomed Pharmacother. 2021;137:111324. doi:10.1016/j.biopha.2021.111324

104. Kharazmkia A, Ziaie S, Ahmadpoor P, et al. A Triple-Blind Randomized Controlled Trial on Impacts of Pioglitazone on Oxidative Stress Markers in Diabetic Kidney Transplant Recipients. Shiraz E-Med J. 2020;21(11). doi:10.5812/semj.98656.

105. Mullur R, Liu -Y-Y, Brent GA. Thyroid Hormone Regulation of Metabolism. Physiol Rev. 2014;94(2):355–382. doi:10.1152/physrev.00030.2013

106. Morte B, Bernal J. Thyroid Hormone Action in the Adult Brain: gene Expression Profiling of the Effects of Single and Multiple Doses of Triiodo-l-Thyronine in the Rat Striatum. Endocrinology. 2008;149(8):3989–4000. doi:10.1210/en.2008-0350

107. Desouza LA, Ladiwala U, Daniel SM, Agashe S, Vaidya RA, Vaidya VA. Thyroid hormone regulates hippocampal neurogenesis in the adult rat brain. Mol Cell Neurosci. 2005;29(3):414–426. doi:10.1016/j.mcn.2005.03.010

108. Lechan RM, Fekete C. Central regulation of hypothalamic-pituitary-thyroid axis under physiological and pathophysiological conditions. Endocr Rev. 2014;35(2):159–194. doi:10.1210/er.2013-1087

109. Chernow B, Burman KD, Johnson DL, et al. T3 may be a better agent than T4 in the critically ill hypothyroid patient. Crit Care Med. 1983;11(2):99–104. doi:10.1097/00003246-198302000-00009

110. Cheng SY. Multiple mechanisms for regulation of the transcriptional activity of thyroid hormone receptors. Reviews in Endocrine and Metabolic. Rev Endocr Metab Disord. 2000;1(12):9–18. doi:10.1023/A:1010052101214

111. Ortiga-Carvalho TM, Sidhaye AR, Wondisford FE. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat Rev Endocrinol. 2014;10(10):582–591. doi:10.1038/nrendo.2014.143

112. Alhowail A. Molecular insights into the benefits of nicotine on memory and cognition (Review). Mol Med Rep. 2021;23(6). doi:10.3892/mmr.2021.12037

113. Leach PT, Kenney JW, Connor DA, Gould TJ. Thyroid receptor β involvement in the effects of acute nicotine on hippocampus-dependent memory. Neuropharmacology. 2015;93:155–163. doi:10.1016/j.neuropharm.2015.01.026

114. Wilcoxon JS, Nadolski GJ, Samarut J, Chassande O, Redei EE. Behavioral inhibition and impaired spatial learning and memory in hypothyroid mice lacking thyroid hormone receptor α. Behav Brain Res. 2007;177(1):109–116. doi:10.1016/j.bbr.2006.10.030

115. McAninch EA, Bianco AC. The history and future of treatment of hypothyroidism. Ann Intern Med. 2016;164(1):50. doi:10.7326/M15-1799

116. Ge JF, Peng L, Hu CM, Wu TN. Impaired learning and memory performance in a subclinical hypothyroidism rat model induced by hemi-thyroid electrocauterisation. J Neuroendocrinol. 2012;24(6):953–961. doi:10.1111/j.1365-2826.2012.02297.x

117. Alzoubi KH, Aleisa AM, Gerges NZ, Alkadhi KA. Nicotine reverses adult-onset hypothyroidism-induced impairment of learning and memory: behavioral and electrophysiological studies. J Neurosci Res. 2006;84(5):944–953. doi:10.1002/jnr.21014

118. Singh S, Rana P, Kumar P, Shankar LR, Khushu S. Hippocampal neurometabolite changes in hypothyroidism: an in vivo h magnetic resonance spectroscopy study before and after thyroxine treatment. J Neuroendocrinol. 2016;28(9):548. doi:10.1111/jne.12399

119. Cooke GE, Mullally S, Correia N, O’Mara SM, Gibney J. Hippocampal volume is decreased in adults with hypothyroidism. Thyroid. 2014;24(3):433–440. doi:10.1089/thy.2013.0058

120. Baghcheghi Y, Salmani H, Beheshti F, et al. Effects of PPAR-γ agonist, pioglitazone on brain tissues oxidative damage and learning and memory impairment in juvenile hypothyroid rats. Int J Neurosci. 2019;129(10):1024–1038. doi:10.1080/00207454.2019.1632843

121. Freche D, Lee C-Y, Rouach N, Holcman D. Synaptic transmission in neurological disorders dissected by a quantitative approach. Commun Integr Biol. 2014;5(5):448–452. doi:10.4161/cib.20818

122. Pereda AE. Electrical synapses and their functional interactions with chemical synapses. Nat Rev Neurosci. 2014;15(4):250–263. doi:10.1038/nrn3708

123. Kennedy MB. Synaptic signaling in learning and memory. Cold Spring Harb Perspect Biol. 2016;8(2):a016824. doi:10.1101/cshperspect.a016824

124. Voglis G, Tavernarakis N. The role of synaptic ion channels in synaptic plasticity. EMBO Rep. 2006;7(11):1104–1110. doi:10.1038/sj.embor.7400830

125. Sudhof TC. Calcium Control of Neurotransmitter Release. Cold Spring Harb Perspect Biol. 2011;4(1):a011353–a011353.

126. Abraham WC, Jones OD, Glanzman DL. Is plasticity of synapses the mechanism of long-term memory storage? Npj Sci Learning. 2019;4(1). doi:10.1038/s41539-019-0048-y

127. Pinto JGA, Jones DG, Murphy KM. Comparing development of synaptic proteins in rat visual, somatosensory, and frontal cortex. Front Neural Circuits. 2013;7. doi:10.3389/fncir.2013.00097

128. Zhao Y, Lützen U, Gohlke P, Jiang P, Herdegen T, Culman J. Neuroprotective and antioxidative effects of pioglitazone in brain tissue adjacent to the ischemic core are mediated by PI3K/Akt and Nrf2/ARE pathways. J Mol Med. 2021;99(8):1073–1083. doi:10.1007/s00109-021-02065-3

129. Liu LP, Yan TH, Jiang LY, et al. Pioglitazone ameliorates memory deficits in streptozotocin-induced diabetic mice by reducing brain β-amyloid through PPARγ activation. Acta Pharmacol Sin. 2013;34(4):455–463. doi:10.1038/aps.2013.11

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.