Skip to main content

Role of GABA pathway in motor and non-motor symptoms in Parkinson's disease: a bidirectional circuit

Abstract

Parkinson's disease (PD) is a progressive neurodegenerative disease as a result of the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The fundamental features of PD are motor and non-motor symptoms. PD symptoms develop due to the disruption of dopaminergic neurotransmitters and other neurotransmitters such as γ-aminobutyric acid (GABA). The potential role of GABA in PD neuropathology concerning the motor and non-motor symptoms of PD was not precisely discussed. Therefore, this review intended to illustrate the possible role of GABA in PD neuropathology regarding motor and non-motor symptoms. The GABA pathway is essential in regulating the inhibitory tone to prevent excessive stimulation of the cerebral cortex. Degeneration of dopaminergic neurons in PD is linked with reducing GABAergic neurotransmission. Decreasing GABA activity promotes mitochondrial dysfunction and oxidative stress, which are highly related to PD neuropathology. Hence, restoring GABA activity by GABA agonists may attenuate the progression of PD motor symptoms. Therefore, dysregulation of GABAergic neurons in the SNpc contributes to developing PD motor symptoms. Besides, PD non-motor symptoms are also related to the dysfunction of the GABAergic pathway, and amelioration of this pathway may reduce PD non-motor symptoms. In conclusion, the deregulation of the GABAergic pathway in PD might be intricate in developing motor and non-motor symptoms. Improving this pathway might be a novel, beneficial approach to control PD symptoms.

Introduction

Parkinson's disease (PD) is a progressive neurodegenerative disease as a result of the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) [1]. PD is the second worldwide neurodegenerative disease after Alzheimer's disease (AD). It affects 1–3% of the population worldwide > 65 years [2]. Notably, two types of PD are well-recognized: familial (genetic) PD and idiopathic (sporadic) PD; familial PD represents 10–15% of all PD types [3]. Many genes contribute to the pathogenesis of PD, including α-synuclein (SNCN), leucine-rich repeat kinase 2 (LRRK2), glucocerebrosidase (GBA), vacuolar protein sorting associated protein 35 (VPS35), phosphatase homolog-induced kinase (PINK1) and Parkinson protein 7 (PAPK7) [3, 4]. The interaction between the susceptible genes and environmental elements influences the onset of PD [5].

Different risk factors, such as sex, age, and ethnicity, contribute to the development of PD [9]. Old age is the leading factor that affects the onset severity of PD. Most PD cases have the age of 60–65 years; however, juvenile PD was reported at the age of less than 21 years [6]. PD is more prevalent in men and white occidental populations [6]. The black race has a lower PD incidence owing to the higher concentration of the neuroprotective neuromelanin [7].

Moreover, heavy metals such as iron, lead and manganese contribute to PD neuropathology via multiple mechanisms, including oxidative stress and mitochondrial dysfunction with subsequent synaptic dysfunction and disruption of brain neurotransmission [8]. Also, drug abuse such as cocaine increases the risk of PD through the induction of the dopaminergic neurons in the SNpc [9] (Fig. 1).

Fig. 1
figure 1

Pathophysiology of PD

Neuronal cell death in the basal ganglion is the primary pathological event in PD. This is attributed to the deposition of α-synuclein [10]. Notably, normal α-synuclein is usually found in the presynaptic site and is significant in releasing neurotransmitters, including dopamine [11]. Genetic environmental factors trigger misfolding and aggregation of α-synuclein and the formation of Lewy bodies [12]. Such changes in neurons could provoke the death of neuronal cells and astrocytes with robust activation of microglia in the SNpc [13]. The accumulated α-synuclein activates degeneration of the dopaminergic neurons in the SNpc either directly due to the toxic impact of α-synuclein or indirectly through induction of mitochondrial dysfunction and proteasomal/lysosomal dysfunctions [14]. Remarkably, the neuropathology of PD occurs first in the olfactory bulb and medulla before affecting the SNpc [15]. The principal neuronal tracts connecting basal ganglia to the other brain regions are orbitofrontal, limbic, associative, oculomotor, and motor tracts are affected in PD neuropathology, causing both motor and non-motor symptoms [16]. Dopamine neurotransmitter released from the dopaminergic neurons is responsible for regulating motor activity. However, low dopamine level is connected with hypokinesia, while increasing dopamine activity leads to dyskinesia, highlighting a defect in motor activity due to dopamine activity and sensitivity [17].

The primary clinical characteristics of PD are motor symptoms such as bradykinesia, postural instability, rigidity, and resting tremors that start when more than 70% of the dopaminergic neurons in the SNpc are damaged [18]. Non-motor symptoms, such as autonomic dysfunction, anosmia, constipation, sleep disorders, and cognitive dysfunction, are usually initiated before the motor symptoms by decades [19]. PD symptoms are correlated to the disturbance of various neurotransmitters, such as dopamine, acetylcholine (Ach), and γ-aminobutyric acid (GABA) [20]. There is little information regarding the role of GABA in PD neuropathology, mainly concerning the development of motor and non-motor symptoms. Thus, this review aimed to clarify the potential role of GABA in PD regarding motor and non-motor symptoms.

GABA overview

GABA is a multi-functional molecule in the CNS, peripheral nervous system (PNS) and non-neuronal tissues [21]. GABA is an inhibitory neurotransmitter extensively expressed in the central nervous system (CNS) [22]. GABA acts on the GABA receptors comprising GABAA, GABAB and GABAC [23]. GABAB is a G-protein metabotropic receptor, while GABAA and GABAC are Cl-gated channels [24] (Fig. 2).

Fig. 2
figure 2

GABA and GABA receptors: glutamate is converted to GABA by the enzyme glutamic acid decarboxylase, which is then stored in the specialized vesicles. The GABA is released upon presynaptic activation of GABAergic neurons, which activates Cl- ion entrance through activation of GABA receptors, leading to hyperpolarization and inhibition of the postsynaptic membrane. Excess GABA in the synaptic cleft is reuptake through GABA transporter to the presynaptic neurons

GABA receptor comprises three central subunits: α, β and γ in a heteromeric or homomeric fashion [22]. GABA regulates neuronal activity through the opening of K+ or inhibiting of Ca+2 via the G-protein signaling pathway [23]. GABA is synthesized from glutamate by the enzyme glutamic acid decarboxylase (GAD); the formed GABA is transported to the presynaptic terminals and stored in specialized vesicles [23]. GAD is widely expressed in the CNS inhibitory neurons and associated with GABA neurons. Dysregulation of GAD is linked with the development of schizophrenia and epilepsy [25].

Depolarization of presynaptic GABergic neurons activates the release of GABA into synaptic space with succeeding stimulation of post-GABA receptors and postsynaptic inhibition [23]. In addition, GABA from the synaptic cleft may leak outside and activate extra-synaptic GABA receptors, causing tonic inhibition [25]. Mainly, GABAA in the dorsal horn is found in presynaptic and postsynaptic neurons, mediating presynaptic inhibition and primary afferent depolarization correspondingly [26]. Extra-synaptic α5GABAA on the proprioceptive afferent neurons leads to tonic depolarization of the spinal cord via modulation of Na+2 channels [26]. Meaningfully, GABAergic neurons are excitatory during prenatal and postnatal periods; however, these neurons undergo developmental changes from excitatory to inhibitory [27]. K + -Cl mainly mediates the polarity of GABA neurons to inhibitory functions− co-transporter (KCC2) [27]. Oxytocin plays a critical role in the transition of GABAergic neurons to inhibitory processes through modulation of KCC2 [28]. In the absence of oxytocin, the activity of KCC2 is reduced with an increase in hyper-excitability state and related disorders like epilepsy and neurodevelopmental disorders [28]. Besides, expressions of GABAA receptors are regulated by allopregnanolone steroid, which exerts positive and negative impacts in acute and chronic effect correspondingly on the expression of GABAA receptors and the progression of dementia [29].

Furthermore, dysfunction of GABAergic neurons in the frontal lobe increases the risk of developing frontotemporal dementia [30]. Restoration of GABAergic neurotransmission by GABA transporter inhibitor tiagabine prevents the development of frontotemporal dementia in both animals and humans [30]. These findings indicated the potential role of GABAergic neurons in regulating neuronal inhibition and preventing neuronal hyper-excitability.

GABA pathway and neurodegenerative disorders

GABAergic neurons are intricate in regulating memory and learning, which are significant variables of AD [31]. Progressive neuronal accumulation of Aβ distorts synaptic function and induces the progression of cognitive impairment through dysregulation of GABAergic neurotransmission [31]. Interaction of Aβ with the neurotransmission network in brain areas linked to memory, such as the hippocampus and amygdaloidal complex, leads to memory impairment and cognitive dysfunction [32]. Regulation of cognitive function is mostly fine-tuning between the excitatory neurotransmitters and GABA inhibitory systems [33]. Hippocampal and cortical functions mainly depend on the GABA inhibitory function to maintain the activity of synaptic plasticity [34]. Hippocampus GABA inhibitory neurons represent 10–15% of total brain inhibitory neurons [34]. It has been revealed that GABAergic neurons are significantly dysregulated and intricate in the pathogenesis of AD [32, 33]. In AD, excitatory neurotransmitters are involved in the pathogenesis of Aβ and tau deposition due to reduced protective GABA inhibitory function [31]. Consequently, disruption of the inhibitory/excitatory axis in the brain could be a possible mechanism for the progress of seizures in AD patients [35].

Indeed, GABAergic neurons are highly distressed in various neurodegenerative diseases, like AD and could be a therapeutic goal in controlling such disorders [31]. A study explained that the density of GABAergic neurons declined next to the Aβ plaques [31], proposing the toxic impacts of Aβ plaques on GABAergic neuron viability. In the AD mice model, it was found that the Aβ plaques are much more harmful to the hippocampal GABAergic neurons compared to other neurons [36]. An in vitro demonstrated that Aβ has differential neurotoxicity for GABAergic neurons [37]. Aβ triggers cell membrane perforation with increasing Ca + 2 effluxes in the hippocampal GABAergic neurons with disturbances of excitatory–inhibitory synaptic function [37]. In AD patients, the concentration of GABA is highly abridged in the temporal cortex and other brain regions [38]. Furthermore, the level of GABA is declined in the cerebrospinal fluid (CSF) in AD [38].

Moreover, deficiency of GABA is associated with the development of Huntington's chorea and other neurodegenerative disorders, as well as neuropsychiatric disorders like panic, depression, and anxiety [39]. Likewise, inflammatory reactions in multiple sclerosis (MS), mainly IL-1β, inhibit GABA function with significant alterations of the GABA pathway [40]. A study observed that CSF GABA declined in MS patients concerning controls [41]. Alteration of GABAergic neurons and reduction of GABA levels are also related to the severity of cognitive deficiency in MS patients [41]. Furthermore, the GABA pathway is highly deregulated in amyotrophic lateral sclerosis (ALS), leading to cortical hyper-excitability [42]. A cross-sectional study revealed reduced GABAergic neurons in ALS patients, causing more severe motor alterations [42].

These findings proposed that the GABA pathway is highly dysregulated in patients with neurodegenerative disorders, and targeting this pathway could be a potential therapeutic strategy against the development and progression of neurodegenerative disorders.

Role of GABA pathway in PD

GABA pathway plays a critical role in regulating inhibitory tone on the globus pallidus (GP), SNpc, and thalamus, preventing excessive stimulation of the cerebral cortex [43]. Dysregulation of the GABA pathway in PD triggers neuronal hyper-excitability, leading to dyskinesia or bradykinesia [43, 44]. Deregulation of the GABA pathway may be involved in developing and progressing motor and non-motor manifestations in PD [44].

GABA and motor manifestations of PD

The mechanism of motor dysfunction in PD is well-defined due to dopaminergic dysfunction; however, alteration of other neurotransmitters like serotonin, Ach, glutamate, noradrenaline, and GABA are also involved in PD neuropathology [45, 46]. GABAergic signaling controls cognition, information processing, and sensory perception [46]. Cardinal motor dysfunction in PD, like resting tremor, bradykinesia, and rigidity, are developed due to the degeneration of dopaminergic neurons in the SNpc. In advanced PD, dyskinesia and motor fluctuation progress due to the degeneration of non-dopaminergic pathways [46].

Disturbance of GABAergic neurons had been reported in the basal ganglia at the postmortem of PD [47]. Tritsch et al. [48] confirmed that dopamine is co-released with GABA from dopaminergic neurons independent of vesicular GABA transporters. The release of GABA also requires the activation of vesicular monoamine transporter 2 (VMAT2), which is also a neurotransmitter of dopamine [48]. Interestingly, the expression of VMAT2 on the GABAergic neurons has an essential role in the release of GABA [49]. Besides, dopaminergic neurons in the SNpc inhibit the striatum via presynaptic activation of GABA receptors [49]. The dopaminergic neurons obtain GABA through presynaptic uptake and then release with dopamine by GABA transporters [49]. Increasing striatal input due to a deficiency of GABA leads to the development of bradykinesia in PD [50]. Selective depletion of dopaminergic receptor 2 (D2R) from an indirect pathway leads to severe motor impairment in mice by decreasing GABAergic neurotransmission [50]. Therefore, D2R is essential for GABAergic neurotransmission and motor control. These observations suggest a mutual interaction between dopamine and GABA. Thus, degeneration of dopaminergic neurons is linked with reducing GABA levels in PD.

It has been reported that the GABA pathway is highly dysregulated in PD neuropathology by disturbing Ca2+ cellular signaling. GABA/Ca2+ maintains neuronal activity in the CNS by preventing intracellular deposits of proteins, Ca2+ and Lewy bodies [51]. Excessive Ca2+ accumulations stimulate α-synuclein aggregation and injury of dopaminergic neurons through induction of excitotoxicity and apoptosis, leading to the development of PD [52]. Notably, the diminution of inhibitory GABAergic neurons triggers the over-activation of cholinergic neuronal signaling, leading to progressive muscle contraction of both agonist and antagonist muscles with the development of stiffness and postural instability [53]. The hyperpolarization of GABAergic neurons regulates presynaptic neurotransmission and prevents neuronal hyper-excitability by maintaining Ca2+ homeostasis [53]. This effect attenuates Ca2+ dyshomeostasis-induced dopaminergic neuron injury. Dopaminergic neurons are highly susceptible to the neurotoxic effects of α-synuclein due to higher expression of Ca2+ voltage-gated channels [54]. Ca2+ voltage-gated channels improve the release of α-synuclein in vitro and in vivo with subsequent aggregation and development of synucleinopathies [54]. Therefore, regulation of Ca2+ voltage-gated channels by GABA may prevent Ca2+-induced excitotoxicity, oxidative stress, mitochondrial dysfunction and development of PD. Reduction of GABA promotes oxidative stress and mitochondrial dysfunction, which is linked with PD neuropathology [55]. Of note, neurosteroids which promote the synthesis of neuronal GABA are highly reduced in PD and other neurodegenerative ailments, leading to excitotoxicity and apoptosis [64]. Experimental studies showed that dysregulation of GABAergic neurons in the SNpc leads to abnormal neuronal firing in rat PD models [65]. A recent study demonstrated that induction of GABAergic neuron generation by astrocyte reprogramming improves motor symptoms in experimental PD [66]. A recent study revealed that presynaptic inhibition in the motor cortex is highly reduced in PD patients, which may explain PD's non-dopaminergic feature [67]. A study exhibited that GABAergic neuron activity in the upper brainstem is reduced compared to the controls [68]. Thus, restoring GABA activity by GABA agonists can attenuate motor symptoms in the PD model [56]. GABA agonists like baclofen and acamprosate protect dopaminergic neurons and striatal terminals from oxidative stress in 6-hydroxydopamine (6-OHDA)-induced PD in rats [56]. Combining baclofen and acamprosate inhibits glutamatergic neurotransmission, improving GABAergic neurotransmission and suppressing neuronal apoptosis and neuroinflammation [56]. A preclinical study observed that GABAB receptor agonist baclofen attenuates motor deficits in MPTP-induced PD in rats by improving GABAergic neurotransmission in the SNpc. Supporting this finding, GABAB receptor antagonist CGP35348 reverses the protective effect of baclofen in MPTP-induced PD in rats [58]. Likewise, Lozovaya et al. [57] showed that the inhibitory effects of GABAergic neurons regulate cholinergic excitatory drive. Consequently, improving inhibitory GABAergic neurons by GABA agonists may reduce the severity of motor symptoms in PD [57]. Bumetanide is an antagonist of chloride importer that improves brain inhibition by reducing intracellular chloride and increases GABAergic neurotransmission in PD patients [59]. Therefore, the augmentation activity of GABAergic neurons may improve motor symptoms in PD. GABAA agonist zolpidem has a remarkably beneficial effect in reducing dyskinesia even after a single dose in PD patients [60]. Zolpidem has a peculiar effect on movement disorders in PD patients, as the use of other GABAA receptor agonist hypnotics like zopiclone and triazolam produced no beneficial motor effects in women with PD [60]. Bohen et al. [61] found that a reduction of GABAA receptor expression in the thalamus correlates with motor dysfunction in PD patients [61]. Conversely, tremor, rigidity, and bradykinesia are developed due to GABAergic neuron hyperactivity driven by GP on the thalamus, and thalamocortical GABAergic neuron activity is increased in PD, as documented in a case–control study [62]. Motor cortex GABA level is inversely correlated to PD disease so that GABA depletion may participate in the development of motor symptoms [62]. However, a case–control study observed that GABA concentration was greater in pons concerning the putamen in the early PD [63], suggesting that altering GABAergic inhibitory tone in the brainstem could be an early neuropathological finding in PD. Therefore, administration of the GABAA receptor antagonist flumazenil has improved postural instability in PD patients [69]. In addition, the GABAA receptor antagonist restores dopaminergic firing and regulates motor dysfunction in PD mouse model [70]. Interestingly, dysregulation of GABAA receptor expression is differentially affected in PD increased in the cerebral cortex and reduced in other brain regions [71], signifying a specific alteration of GABAA receptor expression rather than generalized dysregulation.

These findings suggest that dysregulation of GABAergic neurons in the SNpc contributes to the development of motor symptoms in PD, and targeting this pathway could be a novel approach to managing the motor symptoms of PD.

GABA and non-motor manifestations of PD

Non-motor manifestations in PD, like cognitive dysfunction, sleep disorders, olfactory dysfunction, gastrointestinal disorders, and visual disturbances, represent the primary source of PD burden [72]. These manifestations usually occur many years before the development and progression of motor symptoms. The underlying mechanism for developing these manifestations could be related to GABAergic dysfunction [73]. It has been shown that disturbance of GABAergic neurons contributes to the development of non-motor symptoms in early PD as GABA is co-released with dopamine in the striatum. Thus, the degeneration of the dopaminergic neurons in the SNpc affects GABAergic neurotransmission [74]. Firbank and his colleagues [75] found that GABA concentration was reduced in the occipital cortex, leading to visual hallucination. A cohort study involving 39 PD patients, 19 with hallucination and 17 without hallucination, showed that GABA concentration measured by magnetic resonance spectroscopy was reduced in the occipital cortex and correlated with excitability in PD patients with hallucination [75]. It has been shown that PD patients had visual disturbances with abnormal color vision in the late stage due to alteration of retinal GABAergic neurons [76]. Depletion of retinal GABAergic neurons leads to the development of visual disturbances [77]. Remarkably, increasing retinal GABAergic neurons by GABA agonists also induces visual disturbances [78]. Thus, an optimal GABA level is essential for accuracy and discrimination. GABAergic neurons regulate visual perception; deregulation of GABAergic neurons is engaged with developing visual disturbance in PD [79]. Visual disturbances and retinal abnormalities are observed in PD patients and animals due to the deposition of α-synuclein in the retina [80]. The development of visual hallucination in PD is complex and may be related to anti-PD medications such as anticholinergic benzhexol [111]. Therefore, the management of visual hallucination in PD is not merely associated with the dysregulation of brain GABA since it is related to other neurotransmitters [111].

Indeed, olfactory disorders are common in PD, and more than 90% of PD patients have this disorder [81]. A study illustrated that the olfactory bulb volume was smaller than the matched controls [81]. The reduced volume of the olfactory bulb is correlated with the reduced volume of the putamen and hippocampus [81]. A postmortem study revealed the volume of olfactory bulbs is reduced in PD patients compared to healthy controls [82]. Notably, microstructural changes in the olfactory bulb correlate with dopaminergic neuron dysfunction in the putamen [82]. Olfactory dysfunction in PD is correlated with neuronal loss and structural changes in the nucleus basalis, raphe nuclei, and locus coeruleus [83]. These neuroanatomical changes suggest the involvement of serotonergic, noradrenergic, and cholinergic in olfactory dysfunction [83]. GABAergic neurons in the olfactory pathway regulate odor perception and sensitivity [83]. The development of aberrant GABAergic neurons is associated with olfactory dysfunction in the AD mice model [84]. These findings proposed that dysfunction of GABAergic neurons in PD could be the primary mechanism for developing olfactory dysfunction. In this state, the potentiation of GABAergic neurons may mitigate olfactory dysfunction in PD. A recent experimental study demonstrated that GABA agonist muscimol improves olfactory dysfunction in mouse AD model by regulating presynaptic GABA release and improving of GABAergic neurotransmission in the olfactory system [85]. As well, the development of olfactory dysfunction in PD increases the risk of the development of dementia [86]. GABAergic neurotransmission in the olfactory system modulates glutamatergic neurotransmission in the prefrontal cortex, which is implicated in the pathogenesis of PD [85, 86]. Therefore, early recognition and management of olfactory dysfunction may prevent PD-related complications like dementia. It has been shown that benzodiazepine receptors improve the functional activity of olfactory neurons, and activation of these receptors by GABAA receptor agonists can improve olfactory dysfunction [84]. Activation of the olfactory via low oscillation pulse reduces Aβ accumulation and cognitive impairment in mice by increasing the expression of GABAA receptors [87], suggesting that dysfunction of GABAergic neurotransmission in the olfactory system is implicated in the development of cognitive impairment. However, aberrant expression of the GABAA receptor is involved in the development of cognitive dysfunction in PD mouse model by increasing the accumulation of α-synuclein in early PD. In addition, α-synuclein inhibits the release of GABA in the interneurons, leading to olfactory and cognitive dysfunction in PD mouse model [88].

Furthermore, cognitive dysfunction is frequently associated with PD in about 20–25% [87]. It has been reported that PD patients had a greater risk for the development of dementia and cognitive dysfunction compared to the controls [87]. PD-induced cognitive dysfunction and dementia are developed due to cholinergic deficit, α-synuclein-induced neuronal injury, and dysmetabolism [87, 88]. Single nucleotide polymorphisms (SNPs) RYR2SNP rs10495397 in the Korean population is the most frequent SNP-linked development of cognitive impairment with PD through the development of neuroinflammation [88]. Cognitive dysfunction in PD may develop due to dysregulation of various neurotransmitters like Ach and dopamine in the frontostriatal pathway [89]. Cognitive dysfunction in PD is correlated with both motor and non-motor symptoms [90]. Of note, somatostatin-expressing GABAergic neurons have excitatory effects on the cortical circuits regulating neuronal activity [91].

Therefore, dysfunction of GABAergic neurons is associated with the development of cognitive dysfunction. It has been reported that GAD expression was reduced in PD patients, reducing the neuronal synthesis and release of GABA [92]. Findings from a postmortem study involving 19 PD patients and 19 healthy controls showed that GAD67 expression was decreased in the prefrontal cortex of PD patients compared to controls [92]. Nutt et al. [93] observed that increased expression of the L-amino acid decarboxylase gene by VY-AADC01 improves response to L-dopa therapy in PD patients by enhancing GABA activity. In addition, GABA activity is reduced in PD patients during cognitive stress and stimulation [86].

Furthermore, blunted GABA response to dopamine agonists in PD patients leads to behavioral and cognitive abnormalities [86]. These observations suggest that GABAergic dysfunction in PD is linked with the progression of cognitive dysfunction. Thus, augmentation of GABAergic activity by GABAA agonists could be effective in mitigating cognitive dysfunction in PD. However, the antiepileptic GABA transaminase inhibitor vigabatrin did not improve the cognitive function in epileptic patients [94]. Also, tiagabine, which inhibits GABA transporter, can protect dopaminergic neurons in the SNpc and enhance cognitive function in mouse PD models by inhibiting microglial activation [95]. Moreover, GABA transaminase inhibitor valproate, which is commonly used in the management of epilepsy, its long-term use is associated with the development of cognitive impairment.

Conversely, a recent experimental study found valproate has a neuroprotective role and enhances cognitive function in mice with experimental stroke by increasing the release of GABA and enhancing long-term potentiation. It has been shown that valproate improves cognitive function and attenuates degeneration of the dopaminergic neurons in the SNpc by 50% in rotenone-induced PD through inhibition of histone deacetylase, increasing the accumulation of α-synuclein [96]. However, prolonged use of valproate increases PD risk by inducing progressive degeneration of the dopaminergic neurons in the SNpc by alternating the expression of genes involved in PD neuropathology. A recent clinical trial illustrated that most antiepileptic drugs increase PD risk [97]. Thus, GABA-enhancing drugs have conflicting outcomes on cognitive function and PD risk.

Furthermore, PD neuropathology is associated with sleep disorders, which were reported to be up to 98% in PD patients. Sleep disorders like insomnia, daytime sleepiness, sleep fragmentation, restless leg syndrome and REM behavior disorder (RBD) are frequently developed in the early stage of PD [94]. In addition, sleep disorders adversely affect cognitive function in PD patients [95]. A meta-analysis and systematic review showed that sleep disorders, mainly RBD, are associated with cognitive dysfunction [95]. Remarkably, sleep disorders increase the severity and progression of PD by enhancing the release and deposition, and reducing the clearance of α-synuclein [96]. Sleep disorders in PD are due to the reduced activity of GABAergic neurons [97]. Therefore, activation of the GABAergic pathway by benzodiazepines like nitrazepam could be effective in the management of sleep disorders in PD [98]. A clinical trial indicated that benzodiazepine clonazepam is well-tolerated in improving sleep disorders in PD patients [99]. A scoping review illustrated that clonazepam effectively restored normal sleep in PD patients by enhancing brain GABAergic neurotransmission. Zolpidem is effective for insomnia in PD patients through modulation of the GABAergic pathway; it decreases latency for NREM sleep [99]. It has been reported that zolpidem was very effective in treating insomnia in PD patients owing to its short half-life without daytime sleepiness [100]. A systematic review showed that zolpidem, through potentiation of comprised GABergic neurotransmission, is also effective for PD and other movement disorders. Similarly, a clinical trial showed that daily treatment with benzodiazepine receptor agonist eszopiclone improves sleep quality in PD patients by potentiating brain GABA effects [101]. Therefore, these findings proposed that dysregulation of the GABAergic pathway plays a crucial role in the development of sleep disorders in PD, and activation of the GABA pathway may regulate sleep patterns and attenuate the development of sleep disorders.

Furthermore, dysregulation of the GABAergic pathway is linked with the development of neuropsychiatric disorders like depression and anxiety [100]. Notably, somatostatin-expressing GABAergic neurons are reduced in PD patients with Parkin gene mutation [101]. In the CNS, somatostatin is highly co-localized with GABAergic neurons; it acts as a neuromodulator or co-neurotransmitter and regulates the functional activity of these neurons. CSF somatostatin level reflects the density and activity of GABAergic neurons [102]. Different studies have shown that the CSF somatostatin level was reduced in PD [102, 103]. A recent systematic review and meta-analysis showed that PD neuropathology is associated with functional and structural changes in the neuronal circuits involved in the pathogenesis of anxiety and motor deficits [104]. Likewise, a systematic review and meta-analysis illustrated that depression is found in 38% of PD patients and was more associated with the female sex and GBA1 gene mutation [105]. Also, α-synuclein depression is regarded as independent non-motor symptoms in PD that appear in the early stage and continue throughout the disease duration [105]. Luscher et al. [106] hypothesized that dysfunction of the GABAergic pathway was linked with the development of depressive disorders. A study observed that CSF GABA was low in depressed patients compared to the controls [107]. A systematic review revealed that CSF levels of GABA, somatostatin and brain-derived neurotrophic factor (BDNF) are reduced in patients with depression. Likewise, the reduction of plasma GABA is associated with the development of anxiety disorders [108]. These findings suggest that dysfunction of the GABAergic pathway is related to the development of depression and anxiety in PD. Hence, improvement of brain GABA neurotransmission could be an effective therapeutic strategy in treating neuropsychiatric disorders such as depression and anxiety in PD. It has been stated that the antiepileptic drug levetiracetam enhances neuronal GABA release. A clinical trial disclosed that levetiracetam improves anxiety disorders by enhancing GABA neurotransmission [109]. A double-controlled clinical trial demonstrated that levetiracetam improves cognitive impairment, neuropsychiatric disorders and motor deficits in PD patients. In addition, tiagabine attenuates the development of neuropsychiatric manifestations in experimental rats through the modulation of GABA neurotransmission [110]. These verdicts indicated that deregulation of GABAergic neurotransmission in PD is implicated in the development of neuropsychiatric manifestations such as depression and anxiety. Thus, enhancement of GABAergic neurotransmission by GABA agonists and GABA modulators could be a therapeutic strategy in the management of neuropsychiatric disorders in PD.

Finally, gastrointestinal (GIT) disturbances, including constipation, gastroparesis, nausea, vomiting and hypersalivation, are common in PD due to dysfunction of the enteric nervous system (ENS) and degeneration of the vagus nucleus in the brainstem [108]. The GABAergic pathway regulates intestinal motility and peristaltic reflex [109]. All types of GABA receptors are highly expressed in the GIT and regulate excitatory and inhibitory signaling in the ENS, neuroimmune interaction, and GIT inflammation [109]. GABA receptor agonists can improve GIT disturbances and inflammation in mice [109]. Notoriously, GABA at low concentration exerts an inhibitory effect, while higher concentration leads to an inhibitory effect on the GIT peristaltic activity [110]. GABAA agonist muscimol excites the GIT peristaltic activity blocked by GABAA antagonist bicuculline [110]. Therefore, GABA is regarded as a modulator of colonic peristalsis through modulation of Ach release from enteric neurons [110]. Librium is a well-known benzodiazepine used in the management of gastrointestinal disorders through the activation of GABA signaling in the GIT [111].

These observations indicated that dysfunction of the GABAergic pathway in PD is implicated in the GIT disturbances. Thus, non-motor manifestations in PD are developed due to dysfunction of the GABAergic pathway and amelioration of this pathway may reduce PD severity related to non-motor symptoms.

Also, the miRNAs present at the synapse play a crucial role in the regulation of local synaptic proteins and synapse function. Several miRNAs have been identified to regulate the key proteins of the GABA system in various neurodegenerative diseases. A previous research [112] has revealed a molecular connection between the regulation of GABAergic synapse function by synapse miRNA in AD. However, information on this important aspect needs more research. Taken together, targeting synapse miRNAs to modulate the GABA function could be a novel approach to restoring synapse function in AD and other neuropsychiatric disorders. However, further research is still required to fully understand the association between synaptic miRNAs and GABAergic synapse function.

Taken together, dysregulation of the GABAergic pathway in PD could be involved in the development and progression of motor and non-motor symptoms in PD, and enhancement of this pathway by GABA agonists could be an effective therapeutic modality in the management of PD (Tables 1, 2).

Table 1 Human studies revealing the potential role of GABA and effects of GABA modulators on the motor and non-motor symptoms of PD
Table 2 Animal studies revealing the potential role of GABA and effects of GABA modulators on the motor and non-motor symptoms of PD

Conclusions

PD is characterized by motor and non-motor symptoms developed in response to the disruption of neurotransmitters, including GABA. The GABA pathway is extremely deranged in PD patients. The GABA pathway plays a role in regulating inhibitory tone to prevent excessive stimulation of the cerebral cortex. Degeneration of dopaminergic neurons is associated with reducing GABAergic neurotransmission in PD. Reduction of GABA promotes oxidative stress and mitochondrial dysfunction associated with PD neuropathology. Therefore, restoring GABA activity by GABA agonists can attenuate PD motor symptoms. Hence, dysregulation of GABAergic neurons in the SNpc contributes to developing PD motor symptoms. Also, non-motor symptoms in PD are generated due to dysfunction of the GABAergic pathway and amelioration of this pathway may reduce PD severity related to non-motor symptoms.

The deregulation of the GABAergic pathway in PD might be intricate in developing motor and non-motor symptoms of PD. Enhancing this pathway by GABA agonists could be a new therapeutic modality in managing PD. Targeting the GABA pathway might be a novel therapeutic strategy in managing motor and non-motor manifestations in PD.

Availability of data and materials

All data are available in the manuscript.

References

  1. Marogianni C, Sokratous M, Dardiotis E, Hadjigeorgiou GM, Bogdanos D, Xiromerisiou G. Neurodegeneration and inflammation—an interesting interplay in Parkinson’s disease. Int J Mol Sci. 2020;21(22):8421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Safiri S, Noori M, Nejadghaderi SA, Mousavi SE, Sullman MJ, Araj-Khodaei M, Singh K, Kolahi AA, Gharagozli K. The burden of Parkinson’s disease in the Middle East and North Africa region, 1990–2019: Results from the global burden of disease study 2019. BMC Public Health. 2023;23(1):107.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Cherian A, Divya KP, Vijayaraghavan A. Parkinson’s disease–genetic cause. Curr Opin Neurol. 2023;36(4):292–301.

    Article  PubMed  Google Scholar 

  4. Funayama M, Nishioka K, Li Y, Hattori N. Molecular genetics of Parkinson’s disease: contributions and global trends. J Hum Genet. 2023;68(3):125–30.

    Article  PubMed  Google Scholar 

  5. Yi M, Li J, Jian S, Li B, Huang Z, Shu L, Zhang Y. Quantitative and causal analysis for inflammatory genes and the risk of Parkinson’s disease. Front Immunol. 2023;28(14):1119315.

    Article  Google Scholar 

  6. Zirra A, Rao SC, Bestwick J, Rajalingam R, Marras C, Blauwendraat C, Mata IF, Noyce AJ. Gender differences in the prevalence of Parkinson’s disease. Move Disorders Clin Pract. 2023;10(1):86–93.

    Article  Google Scholar 

  7. Aamodt WW, Willis AW, Dahodwala N. Racial and ethnic disparities in parkinson disease: a call to action. Neurol Clin Pract. 2023;13(2):e200138.

    Article  PubMed  Google Scholar 

  8. Zhao Y, Ray A, Portengen L, Vermeulen R, Peters S. Metal exposure and risk of Parkinson’s disease: a systematic review and meta-analysis. Am J Epidemiol. 2023;192:1207–23.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ferreira C, Almeida C, Tenreiro S, Quintas A. Neuroprotection or neurotoxicity of illicit drugs on Parkinson’s disease. Life. 2020;10(6):86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Alomair BM, Al-Kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, De Waard M, Elekhnawy E, Batiha GE. Is sitagliptin effective for SARS-CoV-2 infection: false or true prophecy? Inflammopharmacology. 2022;30(6):2411–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Al-Kuraishy HM, Al-Gareeb AI, Albogami SM, Jean-Marc S, Nadwa EH, Hafiz AA, Negm WA, Kamal M, Al-Jouboury M, Elekhnawy E, Batiha GE. Potential therapeutic benefits of metformin alone and in combination with sitagliptin in the management of type 2 diabetes patients with COVID-19. Pharmaceuticals. 2022;15(11):1361.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Candelise N, Schmitz M, Thüne K, Cramm M, Rabano A, Zafar S, Stoops E, Vanderstichele H, Villar-Pique A, Llorens F, Zerr I. Effect of the micro-environment on α-synuclein conversion and implication in seeded conversion assays. Transl Neurodegenerat. 2020;9:1–6.

    Google Scholar 

  13. Nadwa EH, Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Albogami SM, Alorabi M, Batiha GE, De Waard M. Cholinergic dysfunction in COVID-19: frantic search and hoping for the best. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(3):453–68.

    Article  CAS  PubMed  Google Scholar 

  14. Al-Kuraishy HM, Al-Gareeb AI, Elewa YH, Zahran MH, Alexiou A, Papadakis M, Batiha GE. Parkinson’s disease risk and hyperhomocysteinemia: the possible link. Cell Mol Neurobiol. 2023;19:1–7.

    Google Scholar 

  15. Gómez-Benito M, Granado N, García-Sanz P, Michel A, Dumoulin M, Moratalla R. Modeling Parkinson’s disease with the alpha-synuclein protein. Front Pharmacol. 2020;23(11):356.

    Article  Google Scholar 

  16. Alotaibi B, El-Masry TA, Elekhnawy E, El-Kadem AH, Saleh A, Negm WA, Abdelkader DH. Aqueous core epigallocatechin gallate PLGA nanocapsules: characterization, antibacterial activity against uropathogens, and in vivo reno-protective effect in cisplatin induced nephrotoxicity. Drug Deliv. 2022;29(1):1848–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Al-Kuraishy HM, Al-Gareeb AI, Alkhuriji AF, Al-Megrin WA, Elekhnawy E, Negm WA, De Waard M, Batiha GE. Investigation of the impact of rosuvastatin and telmisartan in doxorubicin-induced acute cardiotoxicity. Biomed Pharmacother. 2022;1(154): 113673.

    Article  Google Scholar 

  18. Batiha GE, Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E. SIRT1 pathway in Parkinson’s disease: a faraway snapshot but so close. Inflammopharmacology. 2023;31(1):37–56.

    Article  CAS  PubMed  Google Scholar 

  19. Al-kuraishy HM, Al-Gareeb AI, Kaushik A, Kujawska M, Ahmed EA, Batiha GE. SARS-COV-2 infection and Parkinson’s disease: possible links and perspectives. J Neurosci Res. 2022;101(6):952–75.

    Article  Google Scholar 

  20. Abdelaziz A, Sonbol F, Elbanna T, El-Ekhnawy E. Exposure to sublethal concentrations of benzalkonium chloride induces antimicrobial resistance and cellular changes in Klebsiella pneumoniae clinical isolates. Microb Drug Resist. 2019;25(5):631–8.

    Article  CAS  PubMed  Google Scholar 

  21. Al-Kuraishy HM, Hussian NR, Al-Naimi MS, Al-Gareeb AI, Al-Mamorri F, Al-Buhadily AK. The potential role of pancreatic γ-aminobutyric acid (GABA) in diabetes mellitus: a critical reappraisal. Int J Prevent Med. 2021;12:19.

    Google Scholar 

  22. Elekhnawy E, Negm WA. The potential application of probiotics for the prevention and treatment of COVID-19. Egypt J Med Human Genet. 2022;23(1):1–9.

    Article  Google Scholar 

  23. Huang YH, Huang CY. The complexed crystal structure of dihydropyrimidinase reveals a potential interactive link with the neurotransmitter γ-aminobutyric acid (GABA). Biochem Biophys Res Commun. 2022;15(692): 149351.

    Google Scholar 

  24. Mahul-Mellier AL, Burtscher J, Maharjan N, Weerens L, Croisier M, Kuttler F, Leleu M, Knott GW, Lashuel HA. The process of Lewy body formation, rather than simply α-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc Natl Acad Sci. 2020;117(9):4971–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lee SE, Lee Y, Lee GH. The regulation of glutamic acid decarboxylases in GABA neurotransmission in the brain. Arch Pharmacal Res. 2019;42:1031–9.

    Article  CAS  Google Scholar 

  26. Tudeau L, Acuña MA, Albisetti GW, Neumann E, Ralvenius WT, Scheurer L, Poe M, Cook JM, Johannssen HC, Zeilhofer HU. Mice lacking spinal α2GABAA receptors: altered GABAergic neurotransmission, diminished GABAergic antihyperalgesia, and potential compensatory mechanisms preventing a hyperalgesic phenotype. Brain Res. 2022;15(1741): 146889.

    Google Scholar 

  27. Peerboom C, Wierenga CJ. The postnatal GABA shift: a developmental perspective. Neurosci Biobehav Rev. 2021;1(124):179–92.

    Article  Google Scholar 

  28. Li J, Liu H, Guo F, Guo R, Zhang H, He X, Ming X, Ma X, Shang G, Ji P, Song L. Increased GABAergic projections in the paraventricular nucleus regulate colonic hypersensitivity via oxytocin in a rat model of irritable bowel syndrome. NeuroReport. 2023;34(2):108–15.

    Article  CAS  PubMed  Google Scholar 

  29. Bäckström T, Das R, Bixo M. Positive GABAA receptor modulating steroids and their antagonists: implications for clinical treatments. J Neuroendocrinol. 2022;34(2): e13013.

    Article  PubMed  Google Scholar 

  30. Adams NE, Hughes LE, Rouse MA, Phillips HN, Shaw AD, Murley AG, Cope TE, Bevan-Jones WR, Passamonti L, Street D, Holland N. GABAergic cortical network physiology in frontotemporal lobar degeneration. Brain. 2021;144(7):2135–45.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sanchez-Mejias E, Nuñez-Diaz C, Sanchez-Varo R, Gomez-Arboledas A, Garcia-Leon JA, Fernandez-Valenzuela JJ, Mejias-Ortega M, Trujillo-Estrada L, Baglietto-Vargas D, Moreno-Gonzalez I, Davila JC. Distinct disease-sensitive GABAergic neurons in the perirhinal cortex of Alzheimer’s mice and patients. Brain Pathol. 2020;30(2):345–63.

    Article  CAS  PubMed  Google Scholar 

  32. Uemura N, Marotta NP, Ara J, Meymand ES, Zhang B, Kameda H, Koike M, Luk KC, Trojanowski JQ, Lee VM. α-Synuclein aggregates amplified from patient-derived Lewy bodies recapitulate Lewy body diseases in mice. Nat Communicat. 2023;14(1):6892.

    Article  CAS  Google Scholar 

  33. Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GE. Nitazoxanide and COVID-19: a review. Mol Biol Rep. 2022;49(11):11169–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Koh W, Kwak H, Cheong E, Lee CJ. GABA tone regulation and its cognitive functions in the brain. Nat Rev Neurosci. 2023;24(9):523–39.

    Article  CAS  PubMed  Google Scholar 

  35. Vöglein J, Ricard I, Noachtar S, Kukull WA, Dieterich M, Levin J, Danek A. Seizures in Alzheimer’s disease are highly recurrent and associated with a poor disease course. J Neurol. 2020;267:2941–8.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Hidisoglu E, Chiantia G, Franchino C, Marcantoni A, Carbone E. Early Alterations of Hippocampal GABAergic synaptic properties induced by Abeta42 Oligomers. In2020 Alzheimer's Association International Conference 2020. ALZ.

  37. Vazin T, Ball KA, Lu H, Park H, Ataeijannati Y, Head-Gordon T, Poo MM, Schaffer DV. Efficient derivation of cortical glutamatergic neurons from human pluripotent stem cells: a model system to study neurotoxicity in Alzheimer’s disease. Neurobiol Dis. 2014;1(62):62–72.

    Article  Google Scholar 

  38. Govindpani K, Turner C, Waldvogel HJ, Faull RL, Kwakowsky A. Impaired expression of GABA signaling components in the Alzheimer’s disease middle temporal gyrus. Int J Mol Sci. 2020;21(22):8704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sood A, Preeti K, Fernandes V, Khatri DK, Singh SB. Glia: a major player in glutamate–GABA dysregulation-mediated neurodegeneration. J Neurosci Res. 2021;99(12):3148–89.

    Article  CAS  PubMed  Google Scholar 

  40. Batiha GE, Al-Gareeb AI, Elekhnawy E, Al-Kuraishy HM. Potential role of lipoxin in the management of COVID-19: a narrative review. Inflammopharmacology. 2022;30(6):1993–2001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cao G, Edden RA, Gao F, Li H, Gong T, Chen W, Liu X, Wang G, Zhao B. Reduced GABA levels correlate with cognitive impairment in patients with relapsing-remitting multiple sclerosis. Eur Radiol. 2018;28(3):1140–8.

    Article  PubMed  Google Scholar 

  42. Schumacher H, Meyer T, Prüss H. GABAB receptor encephalitis in a patient diagnosed with amyotrophic lateral sclerosis. BMC Neurol. 2019;19(1):1–3.

    Article  Google Scholar 

  43. Alrouji M, Al-Kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, Elekhnawy E, Batiha GE. DPP-4 inhibitors and type 2 diabetes mellitus in Parkinson’s disease: a mutual relationship. Pharmacol Rep. 2023;3:1–4.

    Google Scholar 

  44. Al-kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GE. Dipyridamole and adenosinergic pathway in Covid-19: a juice or holy grail. Egypt J Med Human Genet. 2022;23(1):140.

    Article  Google Scholar 

  45. Al-Kuraishy HM, Al-Fakhrany OM, Elekhnawy E, Al-Gareeb AI, Alorabi M, De Waard M, Albogami SM, Batiha GE. Traditional herbs against COVID-19: back to old weapons to combat the new pandemic. Eur J Med Res. 2022;27(1):186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Blesa J, Foffani G, Dehay B, Bezard E, Obeso JA. Motor and non-motor circuit disturbances in early Parkinson disease: which happens first? Nat Rev Neurosci. 2022;23(2):115–28.

    Article  CAS  PubMed  Google Scholar 

  47. Gerlach M, Gsell W, Kornhuber J, Jellinger K, Krieger V, Pantucek F, Vock R, Riederer P. A post mortem study on neurochemical markers of dopaminergic, GABA-ergic and glutamatergic neurons in basal ganglia-thalamocortical circuits in Parkinson syndrome. Brain Res. 1996;741(1–2):142–52.

    Article  CAS  PubMed  Google Scholar 

  48. Tritsch NX, Ding JB, Sabatini BL. Dopaminergic neurons inhibit striatal output through non-canonical release of GABA. Nature. 2012;490(7419):262–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Melani R, Tritsch NX. Inhibitory co-transmission from midbrain dopamine neurons relies on presynaptic GABA uptake. Cell Rep. 2022;39(3):110716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lemos JC, Friend DM, Kaplan AR, Shin JH, Rubinstein M, Kravitz AV, Alvarez VA. Enhanced GABA transmission drives bradykinesia following loss of dopamine D2 receptor signaling. Neuron. 2016;90(4):824–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Błaszczyk JW. Parkinson’s disease and neurodegeneration: GABA-collapse hypothesis. Front Neurosci. 2016;9(10):269.

    Google Scholar 

  52. Nath S, Goodwin J, Engelborghs Y, Pountney DL. Raised calcium promotes α-synuclein aggregate formation. Mol Cell Neurosci. 2011;46(2):516–26.

    Article  CAS  PubMed  Google Scholar 

  53. Yarnall A, Rochester L, Burn DJ. The interplay of cholinergic function, attention, and falls in Parkinson’s disease. Mov Disord. 2011;26(14):2496–503.

    Article  PubMed  Google Scholar 

  54. Leandrou E, Emmanouilidou E, Vekrellis K. Voltage-gated calcium channels and α-synuclein: implications in Parkinson’s disease. Front Mol Neurosci. 2019;9(12):237.

    Article  Google Scholar 

  55. Ali NH, Alhamdan NA, Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GE. Irisin/PGC-1α/FNDC5 pathway in Parkinson’s disease: truth under the throes. Naunyn Schmiedebergs Arch Pharmacol. 2023;11:1–1.

    Google Scholar 

  56. Hajj R, Milet A, Toulorge D, Cholet N, Laffaire J, Foucquier J, Robelet S, Mitry R, Guedj M, Nabirotchkin S, Chumakov I. Combination of acamprosate and baclofen as a promising therapeutic approach for Parkinson’s disease. Sci Rep. 2015;5(1):1–3.

    Article  Google Scholar 

  57. Lozovaya N, Eftekhari S, Cloarec R, Gouty-Colomer LA, Dufour A, Riffault B, Billon-Grand M, Pons-Bennaceur A, Oumar N, Burnashev N, Ben-Ari Y. GABAergic inhibition in dual-transmission cholinergic and GABAergic striatal interneurons is abolished in Parkinson disease. Nat Commun. 2018;9(1):1–4.

    Article  CAS  Google Scholar 

  58. Tyagi RK, Bisht R, Pant J, Majeed AB, Prakash A. Possible role of GABA-B receptor modulation in MPTP induced Parkinson’s disease in rats. Exp Toxicol Pathol. 2015;67(2):211–7.

    Article  CAS  PubMed  Google Scholar 

  59. Damier P, Hammond C, Ben-Ari Y. Bumetanide to treat Parkinson disease: a report of 4 cases. Clin Neuropharmacol. 2016;39(1):57–9.

    Article  PubMed  Google Scholar 

  60. Daniele A, Panza F, Greco A, Logroscino G, Seripa D. Can a positive allosteric modulation of GABAergic receptors improve motor symptoms in patients with Parkinson’s disease? The potential role of zolpidem in the treatment of Parkinson’s disease. Parkinson’s Disease. 2016;1:2016.

    Google Scholar 

  61. Bohnen NI, Barr J, Vangel R, Roytman S, Paalanen R, Frey KA, Scott PJ, Kanel P. GABAA receptor Benzodiazepine binding sites and motor impairments in Parkinson’s disease. Brain Sci. 2023;13(12):1711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. van Nuland AJ, den Ouden HE, Zach H, Dirkx MF, van Asten JJ, Scheenen TW, Toni I, Cools R, Helmich RC. GABAergic changes in the thalamocortical circuit in Parkinson’s disease. Hum Brain Mapp. 2020;41(4):1017–29.

    Article  PubMed  Google Scholar 

  63. Emir UE, Tuite PJ, Öz G. Elevated pontine and putamenal GABA levels in mild-moderate Parkinson disease detected by 7 tesla proton MRS. PLoS ONE. 2012;7(1): e30918.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Luchetti S, Huitinga IS, Swaab DF. Neurosteroid and GABA-A receptor alterations in Alzheimer’s disease, Parkinson’s disease and multiple sclerosis. Neuroscience. 2011;15(191):6–21.

    Article  Google Scholar 

  65. Wang Y, Zhang QJ, Liu J, Ali U, Gui ZH, Hui YP, Chen L, Wang T. Changes in firing rate and pattern of GABAergic neurons in subregions of the substantia nigra pars reticulata in rat models of Parkinson’s disease. Brain Res. 2010;9(1324):54–63.

    Article  Google Scholar 

  66. Giehrl-Schwab J, Giesert F, Rauser B, Lao CL, Hembach S, Lefort S, Ibarra IL, Koupourtidou C, Luecken MD, Truong DJ, Fischer-Sternjak J. Parkinson’s disease motor symptoms rescue by CRISPRa-reprogramming astrocytes into GABAergic neurons. EMBO Mol Med. 2022;14(5): e14797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Chu J, Wagle-Shukla A, Gunraj C, Lang AE, Chen R. Impaired presynaptic inhibition in the motor cortex in Parkinson disease. Neurology. 2009;72(9):842–9.

    Article  CAS  PubMed  Google Scholar 

  68. Song Y, Gong T, Saleh MG, Mikkelsen M, Wang G, Edden RA. Upper brainstem GABA levels in Parkinson’s disease. Magn Reson Mater Phys Biol Med. 2021;34(5):689–96.

    Article  CAS  Google Scholar 

  69. Ondo WG, Silay YS. Intravenous flumazenil for Parkinson’s disease: a single dose, double blind, placebo controlled, cross-over trial. Mov Disord. 2006;21:1614–7. https://doi.org/10.1002/mds.21022.

    Article  PubMed  Google Scholar 

  70. Emmanouilidou E, Minakaki G, Keramioti MV, Xylaki M, Balafas E, Chrysanthou-Piterou M, Kloukina I, Vekrellis K. GABA transmission via ATP-dependent K+ channels regulates alpha-synuclein secretion in mouse striatum. Brain. 2016;139:871–90. https://doi.org/10.1093/brain/awv403.

    Article  PubMed  Google Scholar 

  71. Zhang W, Xiong BR, Zhang LQ, Huang X, Yuan X, Tian YK, Tian XB. The role of the GABAergic system in diseases of the central nervous system. Neuroscience. 2021;21(470):88–99.

    Article  Google Scholar 

  72. Padilla-Godínez FJ, Ramos-Acevedo R, Martínez-Becerril HA, Bernal-Conde LD, Garrido-Figueroa JF, Hiriart M, Hernández-López A, Argüero-Sánchez R, Callea F, Guerra-Crespo M. Protein misfolding and aggregation: the relatedness between Parkinson’s disease and hepatic endoplasmic reticulum storage disorders. Int J Mol Sci. 2021;22(22):12467.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Jing XZ, Yuan XZ, Luo X, Zhang SY, Wang XP. An Update on Non-dopaminergic treatments for motor and non-motor symptoms of Parkinson’s disease. Curr Neuropharmacol. 2023;21(8):1806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Murueta-Goyena A, Andikoetxea A, Gómez-Esteban JC, Gabilondo I. Contribution of the GABAergic system to non-motor manifestations in premotor and early stages of Parkinson’s disease. Front Pharmacol. 2019;30(10):1294.

    Article  Google Scholar 

  75. Firbank MJ, Parikh J, Murphy N, Killen A, Allan CL, Collerton D, Blamire AM, Taylor JP. Reduced occipital GABA in Parkinson disease with visual hallucinations. Neurology. 2018;91(7):e675–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Haghshomar M, Shobeiri P, Seyedi SA, Abbasi-Feijani F, Poopak A, Sotoudeh H, Kamali A. Cerebellar microstructural abnormalities in Parkinson’s disease: a systematic review of diffusion tensor imaging studies. The Cerebellum. 2022;10:1–27.

    Google Scholar 

  77. Lee JY, Martin-Bastida A, Murueta-Goyena A, Gabilondo I, Cuenca N, Piccini P, Jeon B. Multimodal brain and retinal imaging of dopaminergic degeneration in Parkinson disease. Nat Rev Neurol. 2022;18(4):203–20.

    Article  PubMed  Google Scholar 

  78. Russo M, Carrarini C, Dono F, Rispoli MG, Di Pietro M, Di Stefano V, Ferri L, Bonanni L, Sensi SL, Onofrj M. The pharmacology of visual hallucinations in synucleinopathies. Front Pharmacol. 2019;9(10):1379.

    Article  Google Scholar 

  79. Song XM, Hu XW, Li Z, Gao Y, Ju X, Liu DY, Wang QN, Xue C, Cai YC, Bai R, Tan ZL. Reduction of higher-order occipital GABA and impaired visual perception in acute major depressive disorder. Mol Psychiatry. 2021;26(11):6747–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Veys L, Vandenabeele M, Ortuno-Lizaran I, Baekelandt V, Cuenca N, Moons L, De Groef L. Retinal α-synuclein deposits in Parkinson’s disease patients and animal models. Acta Neuropathol. 2019;137(3):379–95.

    Article  CAS  PubMed  Google Scholar 

  81. Tanik N, Serin HI, Celikbilek A, Inan LE, Gundogdu F. Associations of olfactory bulb and depth of olfactory sulcus with basal ganglia and hippocampus in patients with Parkinson’s disease. Neurosci Lett. 2016;4(620):111–4.

    Article  Google Scholar 

  82. Scherfler C, Esterhammer R, Nocker M, Mahlknecht P, Stockner H, Warwitz B, Spielberger S, Pinter B, Donnemiller E, Decristoforo C, Virgolini I. Correlation of dopaminergic terminal dysfunction and microstructural abnormalities of the basal ganglia and the olfactory tract in Parkinson’s disease. Brain. 2013;136(10):3028–37.

    Article  PubMed  Google Scholar 

  83. Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8(6):329–39.

    Article  CAS  PubMed  Google Scholar 

  84. Chen M, Chen Y, Huo Q, Wang L, Tan S, Misrani A, Jiang J, Chen J, Chen S, Zhang J, Tabassum S. Enhancing GABAergic signaling ameliorates aberrant gamma oscillations of olfactory bulb in AD mouse models. Mol Neurodegener. 2021;16(1):1–23.

    Article  CAS  Google Scholar 

  85. Hu B, Geng C, Guo F, Liu Y, Zong YC, Hou XY. GABAA receptor agonist muscimol rescues inhibitory microcircuit defects in the olfactory bulb and improves olfactory function in APP/PS1 transgenic mice. Neurobiol Aging. 2021;1(108):47–57.

    Article  Google Scholar 

  86. Rodrigues LS, Fagotti J, Targa AD, Noseda AC, Ilkiw JL, Dorieux FW, Lima MM. Olfactory disturbances in Parkinson's disease. InGenetics, Neurology, Behavior, and Diet in Parkinson's Disease 2020 (pp. 539–552). Academic Press.

  87. Svenningsson P, Westman E, Ballard C, Aarsland D. Cognitive impairment in patients with Parkinson’s disease: diagnosis, biomarkers, and treatment. Lancet Neurol. 2012;11(8):697–707.

    Article  PubMed  Google Scholar 

  88. Park KW, Jo S, Kim MS, Jeon SR, Ryu HS, Kim J, Park YM, Koh SB, Lee JH, Chung SJ. Genomic association study for cognitive impairment in Parkinson’s disease. Front Neurol. 2021;4(11): 579268.

    Article  Google Scholar 

  89. Wang W, Mei M, Gao Y, Huang B, Qiu Y, Zhang Y, Wang L, Zhao J, Huang Z, Wang L, Nie K. Changes of brain structural network connection in Parkinson’s disease patients with mild cognitive dysfunction: a study based on diffusion tensor imaging. J Neurol. 2020;267:933–43.

    Article  PubMed  Google Scholar 

  90. Verma G, Bhardwaj A, Aledavood T, De Choudhury M, Kumar S. Examining the impact of sharing COVID-19 misinformation online on mental health. Sci Rep. 2022;12(1):8045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Prévot T, Sibille E. Altered GABA-mediated information processing and cognitive dysfunctions in depression and other brain disorders. Mol Psychiatry. 2021;26(1):151–67.

    Article  PubMed  Google Scholar 

  92. Lanoue AC, Dumitriu A, Myers RH, Soghomonian JJ. Decreased glutamic acid decarboxylase mRNA expression in prefrontal cortex in Parkinson’s disease. Exp Neurol. 2010;226(1):207–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Nutt JG, Curtze C, Hiller A, Anderson S, Larson PS, Van Laar AD, Richardson RM, Thompson ME, Sedkov A, Leinonen M, Ravina B. Aromatic L-amino acid decarboxylase gene therapy enhances levodopa response in Parkinson’s disease. Mov Disord. 2020;35(5):851–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Lajoie AC, Lafontaine AL, Kaminska M. The spectrum of sleep disorders in parkinson disease: a review. Chest. 2021;159(2):818–27.

    Article  PubMed  Google Scholar 

  95. Maggi G, Trojano L, Barone P, Santangelo G. Sleep disorders and cognitive dysfunctions in Parkinson’s disease: a meta-analytic study. Neuropsychol Rev. 2021;31(4):643–82.

    Article  PubMed  Google Scholar 

  96. Wang XT, Yu H, Liu FT, Zhang C, Ma YH, Wang J, Dong Q, Tan L, Wang H, Yu JT. Associations of sleep disorders with cerebrospinal fluid α-synuclein in prodromal and early Parkinson’s disease. J Neurol. 2022;269(5):2469–78.

    Article  CAS  PubMed  Google Scholar 

  97. Radovanovic L, Petrovic J, Saponjic J. Hippocampal and reticulo-thalamic parvalbumin interneurons and synaptic re-organization during sleep disorders in the rat models of Parkinson’s disease neuropathology. Int J Mol Sci. 2021;22(16):8922.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Lim MM, Szymusiak R. Neurobiology of arousal and sleep: updates and insights into neurological disorders. Curr Sleep Med Reports. 2015;1(2):91–100.

    Article  Google Scholar 

  99. Brickley SG, Franks NP, Wisden W. Modulation of GABAA receptor function and sleep. Curr Opin Physio. 2018;1(2):51–7.

    Article  Google Scholar 

  100. Ray S, Agarwal P. Depression and anxiety in Parkinson disease. Clin Geriatr Med. 2020;36(1):93–104.

    Article  PubMed  Google Scholar 

  101. Iwasawa C, Kuzumaki N, Suda Y, Kagawa R, Oka Y, Hattori N, Okano H, Narita M. Reduced expression of somatostatin in GABAergic interneurons derived from induced pluripotent stem cells of patients with parkin mutations. Mol Brain. 2019;12(1):1–8.

    Article  Google Scholar 

  102. Dupont E, Christensen SE, Hansen AP, de Fine OB, Orskov H. Low cerebrospinal fluid somatostatin in Parkinson disease: an irreversible abnormality. Neurology. 1982;32(3):312–4.

    Article  CAS  PubMed  Google Scholar 

  103. Strittmatter M, Hamann GF, Strubel D, Cramer H, Schimrigk K. Somatostatin-like immunoreactivity, its molecular forms and monoaminergic metabolites in aged and demented patients with Parkinson’s disease–effect of L-Dopa. J Neural Transm. 1996;103(5):591–602.

    Article  CAS  PubMed  Google Scholar 

  104. Carey G, Görmezoğlu M, de Jong JJ, Hofman PA, Backes WH, Dujardin K, Leentjens AF. Neuroimaging of anxiety in Parkinson’s disease: a systematic review. Mov Disord. 2021;36(2):327–39.

    Article  PubMed  Google Scholar 

  105. Cong S, Xiang C, Zhang S, Zhang T, Wang H, Cong S. Prevalence and clinical aspects of depression in Parkinson’s disease: a systematic review and meta-analysis of 129 studies. Neurosci Biobehav Rev. 2022;21: 104749.

    Article  Google Scholar 

  106. Luscher B, Shen Q, Sahir N. The GABAergic deficit hypothesis of major depressive disorder. Mol Psychiatry. 2011;16(4):383–406.

    Article  CAS  PubMed  Google Scholar 

  107. Vieira DS, Naffah-Mazacoratti MG, Zukerman E, Soares CS, Alonso EO, Faulhaber MH, Cavalheiro EA, Peres MF. Cerebrospinal fluid GABA levels in chronic migraine with and without depression. Brain Res. 2006;1090(1):197–201.

    Article  CAS  PubMed  Google Scholar 

  108. Skjærbæk C, Knudsen K, Horsager J, Borghammer P. Gastrointestinal dysfunction in Parkinson’s disease. J Clin Med. 2021;10(3):493.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Auteri M, Zizzo MG, Serio R. GABA and GABA receptors in the gastrointestinal tract: from motility to inflammation. Pharmacol Res. 2015;1(93):11–21.

    Article  Google Scholar 

  110. Auteri M, Zizzo MG, Mastropaolo M, Serio R. Opposite role played by GABAA and GABAB receptors in the modulation of peristaltic activity in mouse distal colon. Eur J Pharmacol. 2014;15(731):93–9.

    Article  Google Scholar 

  111. Rossi S, Bartalini S, Ulivelli M. Antiparkinsonian drugs and visual hallucinations. The Lancet Neurology. 2006;1(5):18–9.

    Article  Google Scholar 

  112. Rivera J, Sharma B, Torres MM, Kumar S. Factors affecting the GABAergic synapse function in Alzheimer’s Disease: focus on microRNAs. Ageing Res Rev. 2023;13: 102123.

    Article  Google Scholar 

  113. Liu XY, Wang K, Deng XH, Wei YH, Guo R, Liu SF, Zhu YF, Zhong JJ, Zheng JY, Wang MD, Ye QH. Amelioration of olfactory dysfunction in a mouse model of Parkinson’s disease via enhancing GABAergic signaling. Cell Biosci. 2023;13(1):1–20.

    Article  Google Scholar 

  114. Lai MC, Huang CW. The discordance between network excitability and cognitive performance following vigabatrin treatment during epileptogenesis. Life. 2021;11(11):1213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Liu J, Huang D, Xu J, Tong J, Wang Z, Huang L, Yang Y, Bai X, Wang P, Suo H, Ma Y. Tiagabine protects dopaminergic neurons against neurotoxins by inhibiting microglial activation. Sci Rep. 2015;5(1):15720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. El Sabaa RM, Hamdi E, Hamdy NA, Sarhan HA. Effects of levetiracetam compared to valproate on cognitive functions of patients with epilepsy. Neuropsychiatr Dis Treat. 2020;11:1945–53.

    Article  Google Scholar 

  117. Naseh M, Bayat M, Akbari S, Vatanparast J, Shabani M, Haghighi AB, Haghani M. Neuroprotective effects of sodium valproate on hippocampal cell and volume, and cognitive function in a rat model of focal cerebral ischemia. Physiol Behav. 2022;1(251): 113806.

    Article  Google Scholar 

  118. Monti B, Gatta V, Piretti F, Raffaelli SS, Virgili M, Contestabile A. Valproic acid is neuroprotective in the rotenone rat model of Parkinson’s disease: involvement of α-synuclein. Neurotox Res. 2010;17:130–41.

    Article  CAS  PubMed  Google Scholar 

  119. Brugger F, Bhatia KP, Besag FM. Valproate-associated parkinsonism: a critical review of the literature. CNS Drugs. 2016;30:527–40.

    Article  CAS  PubMed  Google Scholar 

  120. Belete D, Jacobs BM, Simonet C, Bestwick JP, Waters S, Marshall CR, Dobson R, Noyce AJ. Association between antiepileptic drugs and incident Parkinson disease. JAMA Neurol. 2023;80(2):183–7.

    Article  PubMed  Google Scholar 

  121. Hadi F, Agah E, Tavanbakhsh S, Mirsepassi Z, Mousavi SV, Talachi N, Tafakhori A, Aghamollaii V. Safety and efficacy of melatonin, clonazepam, and trazodone in patients with Parkinson’s disease and sleep disorders: a randomized, double-blind trial. Neurol Sci. 2022;43(10):6141–8.

    Article  PubMed  Google Scholar 

  122. de Almeida CM, Pachito DV, Sobreira-Neto MA, Tumas V, Eckeli AL. Pharmacological treatment for REM sleep behavior disorder in Parkinson disease and related conditions: a scoping review. J Neurol Sci. 2018;15(393):63–8.

    Article  Google Scholar 

  123. Vadukul DM, Papp M, Thrush RJ, Wang J, Jin Y, Arosio P, Aprile FA. α-Synuclein aggregation is triggered by oligomeric amyloid-β 42 via heterogeneous primary nucleation. J Am Chem Soc. 2023;145(33):18276–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Bomalaski MN, Claflin ES, Townsend W, Peterson MD. Zolpidem for the treatment of neurologic disorders: a systematic review. JAMA Neurol. 2017;74(9):1130–9.

    Article  PubMed  Google Scholar 

  125. Menza M, Dobkin RD, Marin H, Gara M, Bienfait K, Dicke A, et al. Treatment of insomnia in Parkinson’s disease: a controlled trial of eszopiclone and placebo. Mov Disord. 2010;25:1708–14. https://doi.org/10.1002/mds.23168.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Mousten IV, Sørensen NV, Christensen RH, Benros ME. Cerebrospinal fluid biomarkers in patients with unipolar depression compared with healthy control individuals: a systematic review and meta-analysis. JAMA Psychiat. 2022;79:571–81.

    Article  Google Scholar 

  127. Nasir M, Trujillo D, Levine J, Dwyer JB, Rupp ZW, Bloch MH. Glutamate systems in DSM-5 anxiety disorders: their role and a review of glutamate and GABA psychopharmacology. Front Psychiat. 2020;11:548505.

    Article  Google Scholar 

  128. Stein MB, Ravindran LN, Simon NM, Liebowitz MR, Khan A, Brawman-Mintzer O, et al. Levetiracetam in generalized social anxiety disorder: a double-blind, randomized controlled trial. J Clin Psychiatry. 2010;71:627–31. https://doi.org/10.4088/JCP.08m04949gre.

    Article  CAS  PubMed  Google Scholar 

  129. Dissanayaka N, Pourzinal D, Byrne GJ, Yang J, McMahon KL, Pontone GM, O’Sullivan JD, Adam R, Littleford R, Chatfield M, Lehn A. Levetiracetam for the treatment of mild cognitive impairment in Parkinson’s disease: a double-blind controlled proof-of-concept trial protocol. Pilot Feasibility Stud. 2023;9(1):189.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Dhir A, Akula KK, Kulkarni SK. Tiagabine, a GABA uptake inhibitor, attenuates 3-nitropropionic acid-induced alterations in various behavioral and biochemical parameters in rats. Prog Neuropsychopharm Biol Psychiatry. 2008;32(3):835–43.

    Article  CAS  Google Scholar 

Download references

Funding

Open Access funding enabled and organized by Projekt DEAL. Open Access funding enabled and organized by Projekt DEAL. This work was supported by the University of Witten-Herdecke Germany.

Author information

Authors and Affiliations

Authors

Contributions

HMA, AIA, EE, and GEB contributed to the study conception and design. Material preparation, data collection and analysis, and writing the first draft was performed by HMA, AIA, EE, and GEB. BA, HA, AA, and MP revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Engy Elekhnawy, Marios Papadakis or Gaber El-Saber Batiha.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alharbi, B., Al-kuraishy, H.M., Al-Gareeb, A.I. et al. Role of GABA pathway in motor and non-motor symptoms in Parkinson's disease: a bidirectional circuit. Eur J Med Res 29, 205 (2024). https://doi.org/10.1186/s40001-024-01779-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-024-01779-7

Keywords