Loading

Review Article Open Access
Volume 6 | Issue 2 | DOI: https://doi.org/10.33696/Signaling.6.134

Monoamine Oxidase B in Astrocytic GABA Synthesis: A Central Mechanism in Neurodegeneration and Neuroinflammation

  • 1Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
  • 2Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
+ Affiliations - Affiliations

*Corresponding Author

Moawiah M Naffaa, Moawiah.naffaa@duke.edu

Received Date: February 10, 2025

Accepted Date: April 09, 2025

Abstract

Monoamine oxidase B (MAO-B) is a mitochondrial enzyme predominantly expressed in astrocytes, where it plays a crucial role in neurotransmitter metabolism, oxidative stress regulation, and neuroinflammation. In addition to its well-characterized function in the oxidative deamination of monoamines such as dopamine, noradrenaline, and serotonin, MAO-B is increasingly recognized for its involvement in astrocytic GABA synthesis. In reactive astrocytes, upregulated MAO-B activity enhances GABA production, leading to excessive tonic inhibition that disrupts neuronal excitability and synaptic function. Moreover, MAO-B-mediated metabolism generates reactive oxygen species (ROS), including hydrogen peroxide, further exacerbating oxidative stress and neuroinflammation. This article examines the critical role of MAO-B in neurological disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and stroke, emphasizing its contribution to impaired GABAergic signaling and neurodegeneration. Finally, it explores the potential of MAO-B inhibition as a targeted therapeutic approach to regulate astrocytic GABA synthesis, mitigate aberrant inhibitory signaling, and restore neural circuit homeostasis in neurodegenerative and metabolic disorders.

Keywords

Monoamine oxidase B, Astrocytes, GABAergic signaling, Alzheimer's disease (AD), Parkinson’s disease (PD), Stroke and CNS injury, Reactive oxygen species (ROS), Astrocytic GABA synthesis, Neural circuit, NSCs proliferation activity

Introduction

Monoamine oxidase (MAO) is a mitochondrial enzyme responsible for the oxidative deamination of biogenic amines, including dopamine (DA), serotonin, norepinephrine, and melatonin [1,2]. Through its regulation of neurotransmitter levels, MAO plays a pivotal role in mood regulation, cognition, and motor control [3,4]. MAO exists in two isoforms, MAO-A and MAO-B, which differ in their substrate specificity, tissue distribution, and physiological functions [5,6].

MAO-A primarily metabolizes serotonin, norepinephrine, epinephrine, and melatonin and is predominantly expressed in dopaminergic axon terminals, particularly within the nigrostriatal pathway [1,2,7]. In contrast, MAO-B primarily degrades phenylethylamine and benzylamine, with a secondary role in dopamine metabolism. Notably, MAO-B is highly expressed in astrocytes, where it contributes to neurotransmitter homeostasis in non-neuronal brain cells [8,9]. While both isoforms are capable of metabolizing dopamine, MAO-A is the principal enzyme responsible for dopamine degradation in neurons, due to the high expression of the dopamine transporter (DAT) in dopaminergic terminals. Conversely, MAO-B’s role in dopamine metabolism is limited by the lack of DAT expression in astrocytes, restricting dopamine uptake in these cells [10,11].

Beyond its role in dopamine metabolism, MAO-B plays a crucial role in astrocytic GABA synthesis. In particular, reactive astrocytes modulate neuronal excitability through GABA production. MAO-B facilitates this process by metabolizing putrescine into 4-aminobutanal, which is subsequently converted into GABA [12,13]. This pathway contributes to tonic inhibition of neuronal activity, particularly in the striatum and substantia nigra pars compacta (SNpc). Dysregulation of this process has been implicated in neurodegenerative diseases such as Parkinson’s disease (PD), where reactive astrocytes increase GABA synthesis in response to neuronal injury, leading to excessive tonic inhibition and exacerbating motor deficits [14-17].

In addition to PD, MAO-B has been linked to Alzheimer’s disease (AD) and stroke-related neuronal injury. In AD, elevated MAO-B expression in astrocytes is associated with increased oxidative stress, amyloid-beta aggregation, and neuroinflammation, contributing to cognitive decline [18,19]. Similarly, in ischemic stroke, increased MAO-B activity has been implicated in excitotoxicity and neuronal apoptosis, as reactive astrocytes upregulate MAO-B expression following ischemic events [9]. Consequently, inhibition of MAO-B has been proposed as a therapeutic strategy to mitigate oxidative stress, neuroinflammation, and excessive GABA-mediated tonic inhibition in neurodegenerative and ischemic disorders [20,21].

In addition to its role in astrocytes, lateral ventricle (LV) neural stem cells (NSCs) also play a significant role in GABA regulation. These cells either directly synthesize GABA or uptake it from the extracellular space [22]. Upon activation of the ACC- subependymal -ChAT+ (subep-ChAT+) circuit, which modulates the proliferative activity of LV NSCs, these cells exhibit an upregulation of both MAO-B activity and GABA production [22,23]. This suggests that LV NSCs may synthesize GABA through the MAO-B enzyme, similarly to astrocytes, given that LV NSCs exhibit glial-like behavior. The synthesized GABA is then released into the microenvironment between LV NSCs and subep-ChAT+ neurons. This release is thought to function as a feedback mechanism, potentially modulating the activity of the ACC-subep-ChAT+ circuit and influencing LV NSC proliferation [24]. Such regulatory mechanisms are essential for neurogenesis, as they help balance stem cell activity over time, allowing LV NSCs to exert control over external regulatory influences and maintain homeostasis within the neurogenic niche.

This article investigates the critical role of MAO-B in astrocytic GABA synthesis and its broader implications for neurological health and disease. It examines how MAO-B-mediated metabolism influences GABAergic signaling, particularly in reactive astrocytes, where its dysregulation contributes to excessive GABA production, neurotransmitter imbalance, and neuronal inhibition. Furthermore, the article explores the impact of MAO-B activity in neurodegenerative conditions such as AD, PD, and stroke, emphasizing the potential of MAO-B inhibition as a therapeutic approach to modulate astrocytic function, restore synaptic homeostasis, and mitigate neurodegeneration.

MAO-B in Neurophysiology and Disease: Astrocytic Role in Neurotransmitter Homeostasis, Oxidative Stress, and Therapeutic Implications

The role of MAO-B in astrocytic function, neurotransmitter regulation, and neurological disease pathophysiology

MAO-B is a mitochondrial enzyme that plays a pivotal role in maintaining neurotransmitter homeostasis through the oxidative deamination of monoamines [25,26]. Originally identified in the 1920s as tyramine oxidase in the liver, MAO-B was later reclassified to distinguish it from other oxidative deaminases. By the 1950s, its presence in the brain was established, revealing its capacity to oxidize catecholamines, including DA, adrenaline, and noradrenaline [1,27,28]. These discoveries emphasized the enzyme's critical role in neurophysiology and its broader implications for neural function and disease.

MAO-B is widely distributed in key brain regions, including the cerebral cortex, cerebellum, hippocampus, and midbrain, with its predominant localization in astrocytes. Immunohistochemical and in situ hybridization studies have confirmed its enrichment in astrocytes in both rodent and human brains. It is also present in serotonergic and histaminergic neurons, particularly in the dorsal raphe and tuberomammillary nucleus [29-32]. The predominant presence of MAO-B in astrocytes underscores its essential role in glial-mediated modulation of neurotransmission, regulation of neuroinflammation, and oxidative stress, all of which influence neuronal survival and synaptic function [33-35].

In addition to its role in neurotransmitter regulation, MAO-B is involved in the conversion of polyamines, such as putrescine, into gamma-aminobutyric acid (GABA), a key inhibitory neurotransmitter in the central nervous system (CNS) [17]. This process becomes especially important within reactive astrocytes—glial cells that undergo morphological and functional changes in response to persistent metabolic stress. The MAO-B-mediated synthesis of GABA establishes a critical link between lipid metabolism and neurochemical signaling in the brain, highlighting its relevance in brain function and pathology [36,37].

Astrocytes exposed to chronic levels of medium-chain fatty acids (MCFAs), such as dopamine, exhibit an increase in GABA production, which serves to inhibit neuronal activity [14,38]. This upregulation of GABA synthesis is especially pronounced in brain regions such as the hypothalamus, a central hub for regulating energy homeostasis, feeding behavior, and metabolic balance [39]. Alterations in GABAergic signaling in the hypothalamus may influence broader physiological processes, emphasizing the importance of astrocytic regulation in maintaining brain and body homeostasis [39,40].

Furthermore, MAO-B activation in astrocytes contributes to the generation of reactive oxygen species (ROS), including hydrogen peroxide (H2O2), which induces oxidative stress [34,41]. The accumulation of ROS exacerbates cellular dysfunction by disrupting the balance within astrocytic populations, promoting hypertrophy, proliferation, and inflammation [42]. These changes impair the supportive functions of astrocytes, further compromising neuronal integrity. The interplay between oxidative stress and metabolic reprogramming in reactive astrocytes highlights the complexity of glial responses to chronic metabolic disturbances [43].

Such disturbances are commonly observed in various neurological diseases, where the interplay between GABA synthesis, ROS production, and altered astrocytic function plays a significant role in disease progression [13,44]. These processes may not only contribute to the pathophysiology of neurological disorders but also offer potential therapeutic targets for modulating astrocytic activity and restoring metabolic homeostasis in the brain.

Gliotransmission and astrocytic GABA: Implications for brain homeostasis and neurological disorders

Recent advancements in gliotransmission have revolutionized our understanding of astrocytes, which were once viewed as passive support cells but are now recognized as active participants in neurotransmitter release and neuronal communication [45]. Gliotransmission, the release of neurotransmitters such as GABA and glutamate by astrocytes, plays a pivotal role in maintaining brain function and homeostasis [46-48].

A breakthrough in this field was the identification of a channel-mediated mechanism that allows astrocytes to release GABA, the primary inhibitory neurotransmitter in the CNS, particularly in the cerebellum. The Best1 channel was found to facilitate this release, establishing that astrocytes contribute to tonic GABA inhibition in the cerebellum, a region crucial for motor control [49-51]. This discovery highlighted how astrocytes modulate neuronal activity, maintaining a balance between excitation and inhibition within cerebellar circuits, which is essential for motor coordination [52,53].

Further research uncovered that GABA biosynthesis in astrocytes involves the putrescine degradation pathway, with MAO-B playing a key role in converting putrescine to GABA [17]. This previously unrecognized pathway underscores the adaptability of astrocytes in regulating neurotransmitter availability and neuronal activity, reinforcing their central role in brain homeostasis.

Astrocytes' tonic GABA release is crucial for maintaining inhibitory control over neuronal activity, particularly in the cerebellum, where precise motor control is necessary for coordinated movement. Disruptions in this balance can lead to motor deficits, such as cerebellar ataxia [53,54], highlighting the importance of astrocytic GABA in both normal and pathological motor function.

The pathological role of astrocytic GABA has been explored in various neurodegenerative diseases, including AD, PD, Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) [55,56]. In AD, aberrant GABA releases in the hippocampus impairs memory formation, while in PD, dysregulated release disrupts basal ganglia function, leading to motor deficits. Similar alterations in HD and ALS contribute to motor dysfunction and cognitive decline [57-59]. These findings suggest that astrocytic GABA is crucial for healthy brain function and may serve as a promising biomarker for diseases such as AD and PD. Targeting pathways like MAO-B and Best1 offers potential therapeutic strategies.

Beyond AD and PD, modulating astrocytic GABA may have therapeutic benefits for other neurological conditions. In white matter stroke, astrocytes may influence the maladaptive response to ischemic injury, while in spinal cord injury, GABA release may impact recovery by modulating plasticity-related circuits [60,61]. In metabolic disorders like obesity, dysregulated astrocytic neurotransmitter release can alter circuits involved in appetite and energy balance [62,63]. Additionally, in epilepsy, disruptions in GABA release can exacerbate seizures, underscoring the importance of GABAergic signaling in controlling neural excitability [64,65].

As research advances, it may open new avenues for restoring astrocytic GABA balance in neurological disorders. Targeting molecular pathways that regulate GABA release, such as MAO-B and Best1, could help restore proper GABAergic signaling, offering therapeutic potential for patients with neurodegenerative diseases, epilepsy, and other conditions. These insights highlight the promising future of therapeutic interventions aimed at modulating astrocytic function to restore neural circuit balance and alleviate symptoms in a variety of neurological disorders.

The varying impact of MAO-B expression across different neurological conditions, particularly the relationship between its upregulation and the resulting pathological consequences, is discussed in Table 1. In addition to cognitive decline and neurodegeneration in aging and AD, increased MAO-B expression in regions such as the striatum, substantia nigra, and cortex is directly associated with oxidative stress, excitotoxicity [66,67], and impaired neuronal function in conditions like PD, stroke, and traumatic brain injury (TBI). In diseases such as multiple sclerosis (MS) and ALS, elevated MAO-B expression in astrocytes and microglia promotes neuroinflammation and oxidative damage, resulting in axonal loss, compromised myelin integrity, and motor dysfunction (Table 1) [68,69]. The overexpression of MAO-B in these conditions suggests that its activity may be a key contributor to the progression of neurodegenerative diseases [9,70,71] and a promising avenue for therapeutic intervention.

Table 1. Impact of MAO-B Expression in Various Neurological Conditions.

Condition

Brain Regions Affected

Role of Monoamine Oxidase B (MAO-B)

Impact

References

Aging

Cortex, hippocampus, striatum

Increased expression in astrocytes

Cognitive decline, oxidative stress, and early neurodegeneration

[72,73]

Alzheimer's Disease (AD)

Hippocampus, frontal cortex

Elevated in reactive astrocytes

Neuroinflammation, synaptic dysfunction, and neuronal loss

[18,74-76]

Parkinson's Disease (PD)

Substantia nigra pars compacta

Increased oxidative stress, elevated in astrocytes

Dopaminergic neuron degeneration, exacerbated neuroinflammation

[34,77-79]

Stroke

Peri-infarct regions

Upregulated in response to hypoxia and ischemia

Blood-brain barrier disruption, excitotoxicity, and neuronal injury

[80,81]

Traumatic Brain Injury (TBI)

Perilesional astrocytes

Increased MAO-B activity, prolonged neuroinflammatory response

Prolonged neuroinflammation, exacerbated neuronal damage

[82,83]

Multiple Sclerosis (MS)

Corticospinal tract, cerebellum, optic nerve

Elevated in reactive astrocytes and microglia

Neuroinflammation, axonal damage, and loss of myelin integrity

[84-86]

Amyotrophic Lateral Sclerosis (ALS)

Motor cortex, spinal cord

Elevated in astrocytes and microglia, promoting oxidative stress

Motor neuron degeneration, muscle weakness, and loss of motor function

[87,88]

Huntington's Disease (HD)

Striatum, cortex, globus pallidus

Overexpression in astrocytes, promoting neurodegenerative processes

Striatal atrophy, motor dysfunction, and cognitive decline

[89]


The following sections will discuss the multifaceted role of MAO-B in neurodegenerative diseases and CNS injury, emphasizing its contributions to oxidative stress, neuroinflammation, and neurotransmitter dysregulation. Particular attention will be given to its involvement in Parkinson’s disease and Alzheimer’s disease, where MAO-B activity disrupts dopaminergic and GABAergic signaling, exacerbating disease progression. Additionally, its role in CNS injury will be examined, focusing on how MAO-B-driven H2O2 production promotes astrocyte reactivity and glial scar formation, which, while protective, also impairs neuronal recovery.

MAO-B in cellular metabolism: Implications for glycolysis and astrocytic GABA synthesis

MAO-B is recognized for its role in the oxidative deamination of neurotransmitters and biogenic amines. Although it is not directly involved in glycolysis, emerging evidence suggests that MAO-B activity influences metabolic pathways that intersect with glycolytic processes and cellular energy homeostasis [14,90]. This connection is particularly relevant in astrocytes, where MAO-B significantly contributes to GABA synthesis.

Located on the outer mitochondrial membrane, MAO-B catalyzes the breakdown of amines, generating H2O2 as a byproduct. The accumulation of H2O2 can induce oxidative stress, impair mitochondrial function, and reduce ATP production [91]. In response, cells may upregulate glycolysis as a compensatory mechanism to sustain energy production [92,93]. In astrocytes, such metabolic shifts can profoundly affect neurotransmitter biosynthesis, especially in the conversion of glutamate to GABA [94,95].

Beyond its direct oxidative role, MAO-B activity also impacts cellular redox signaling, which can, in turn, influence glucose metabolism. H2O2 has been shown to activate hypoxia-inducible factor 1-alpha (HIF-1α), a key transcription factor that regulates the expression of glycolytic enzymes [41]. The upregulation of HIF-1α promotes increased glucose uptake and enhances glycolytic flux [96,97], suggesting a potential metabolic adaptation in astrocytes under oxidative stress. This shift may alter the availability of metabolic intermediates required for GABA synthesis, potentially disrupting the balance of inhibitory neurotransmission.

In neurodegenerative diseases such as Alzheimer's and Parkinson's, elevated MAO-B activity, oxidative stress, and significant metabolic alterations in astrocytes are commonly observed [9,34,71,98]. The increased expression of glycolytic enzymes in these conditions points to a reprogramming of astrocytic metabolism [43,99], potentially as a direct consequence of MAO-B activity or as an adaptive response to mitochondrial dysfunction. This metabolic reprogramming may disturb the delicate balance of neurotransmitter synthesis, including astrocytic GABA production, further exacerbating disease pathology.

Regulation of oxidative stress by MAO-B and H2O2: Implications for neurodegenerative diseases

H2O2, produced by MAO-B, plays a critical role in regulating oxidative stress by triggering a cascade of cellular defense mechanisms [100]. This includes the upregulation of key antioxidant enzymes such as catalase, superoxide dismutase (SOD), and peroxidase, which collectively work to neutralize oxidative damage and maintain redox balance [101]. Astrocytes, known for their robust antioxidant capacity, serve as first-line responders to oxidative stress by enhancing the expression and activity of these enzymes [94]. This protective mechanism is essential for shielding neurons from oxidative damage and preserving synaptic integrity [102].

However, under pathological conditions such as Alzheimer’s and Parkinson’s disease, chronic MAO-B activation leads to excessive and sustained H2O2 production, along with other ROS [6,103]. Over time, this persistent oxidative burden overwhelms the cellular antioxidant defense system, resulting in mitochondrial dysfunction, lipid peroxidation, protein aggregation, and DNA damaged hallmark features of neurodegenerative disease pathology [104]. As oxidative stress intensifies, neuronal function becomes increasingly compromised, accelerating disease progression and cognitive decline [105].

Targeting MAO-B represents a promising therapeutic strategy for mitigating oxidative stress and restoring neuronal homeostasis. Pharmacological inhibitors such as selegiline and rasagiline effectively reduce H2O2 production [106], thereby alleviating oxidative stress and preventing cellular damage. Additionally, MAO-B inhibition has been shown to enhance endogenous antioxidant enzyme activity, reinforcing the brain’s natural defense against oxidative injury [1,107]. By reducing neurotoxicity and preserving neuronal integrity, these inhibitors offer a multifaceted approach to combating neurodegenerative diseases, potentially slowing disease progression and improving clinical outcomes.

MAO-B Inhibitors in PD: Mechanisms, Dopamine Metabolism, and Astrocytic Neuroinflammation

PD is a prevalent neurodegenerative disorder characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra, leading to reduced dopamine levels in the striatum [108,109]. This depletion manifests as hallmark motor symptoms, including bradykinesia, resting tremors, rigidity, gait disturbances, and postural instability [110-112]. While levodopa remains the primary treatment, MAO-B inhibitors, such as selegiline and rasagiline, are employed as monotherapy or adjunctive therapy [113,114]. However, their clinical efficacy remains inconsistent, highlighting the need for a deeper understanding of their role in PD pathophysiology.

Although MAO-B’s precise function in dopamine degradation is still under investigation, the therapeutic benefits of its inhibitors suggest a broader role in dopamine regulation. Emerging evidence indicates that these inhibitors primarily exert their effects by reducing tonic inhibition of dopaminergic neurons in the substantia nigra pars compacta rather than directly blocking dopamine metabolism [9,115,116]. Beyond dopamine regulation, MAO-B inhibitors may also confer neuroprotection by stabilizing mitochondria, inducing anti-apoptotic proteins, and promoting neurotrophic factors, potentially mitigating further dopaminergic neuron degeneration [114,117,118]. These findings underscore the need to reassess the mechanisms underlying MAO-B inhibition in PD to refine therapeutic strategies.

Comparative roles of MAO-A and MAO-B in dopamine metabolism and PD

Each isoform of MAOs, MAO-A and MAO-B, exhibits distinct substrate specificity, cellular distribution, and physiological roles. MAO-A is primarily localized in dopaminergic axon terminals, where it plays a key role in degrading epinephrine, norepinephrine, serotonin, and melatonin (Table 2) [2,9,25,119]. In contrast, MAO-B is predominantly expressed in astrocytes and serotonergic neurons, preferentially metabolizing phenylethylamine and benzylamine [8,120,121]. Although both isoforms contribute to dopamine (DA) metabolism, emerging evidence suggests that MAO-A is the principal enzyme responsible for DA degradation [2,122]. These finding challenges previous assumptions about MAO-B’s role in PD and underscores the necessity of re-evaluating MAO-targeted therapeutic strategies.

Early pharmacological studies suggested MAO-B as a key regulator of DA metabolism, but genetic and biochemical analyses have refined this perspective. Studies on MAO-B-deficient mice show no significant alterations in striatal DA levels, whereas selective MAO-A inhibition markedly reduces DA metabolism [123,124]. Additionally, MAO-B inhibitors such as selegiline and rasagiline exert minimal effects on DA efflux in dopaminergic brain regions, reinforcing the role of MAO-A as the primary enzyme in DA catabolism [9,114,116,117].

Beyond DA metabolism, MAO-B is critical for astrocytic function and neuroinflammation. It facilitates the synthesis of astrocytic GABA through putrescine degradation, contributing to excessive tonic inhibition of dopaminergic neurons in the substantia nigra pars compacta, a mechanism associated with motor dysfunction in PD [125-127]. Increased MAO-B expression correlates with reactive astrogliosis, further implicating it in disease progression. The therapeutic benefits of MAO-B inhibitors may therefore stem from reducing astrocytic GABA synthesis and neuroinflammation rather than directly preserving DA levels (Table 2) [9,128,129].

Table 2. Comparative Functional and Pathophysiological Roles of MAO-A and MAO-B in Dopamine Metabolism and Neurodegenerative Diseases.

Feature

MAO-A

MAO-B

References

Primary Location

Dopaminergic axon terminals

Astrocytes, serotonergic neurons

[30,130]

Key Substrates

Dopamine, serotonin, norepinephrine, melatonin

Phenylethylamine, benzylamine

[2,131]

Role in DA Metabolism

Major contributor to DA degradation

Limited involvement in DA degradation

[2,9]

Impact of Inhibition

Reduces DA metabolism and turnover

No significant effect on DA efflux

[132,133]

Relevance to PD

Directly affects DA availability

Implicated in astrocytic GABA synthesis and neuroinflammation

[2,9,126]

Expression Regulation

Modulated by hormonal and environmental factors

Upregulated in aging and neurodegeneration

[120,134,135]

Role in Oxidative Stress

Generates hydrogen peroxide (H2O2)

Contributes to oxidative stress in astrocytes

[34,136]

Association with Neurodegenerative Diseases

Implicated in depression, anxiety, PD

Involved in PD, Alzheimer’s, and neuroinflammation

[121,130]

Role in MPTP Neurotoxicity

Minimal involvement

Convert MPTP into toxic MPP+, damaging DA neurons

[9,137]

Neurotransmitter Turnover Rate

Rapid metabolism of monoamines

Slower turnover, mainly in glial cells

[124,138]

Therapeutic Implications

Targeted inhibition increases synaptic DA

Inhibition reduces astrocytic GABA overproduction

[139,140]

Pharmacological Inhibitors

Clorgyline, moclobemide

Selegiline, rasagiline

[141]


MAO-B is also involved in neurotoxin-induced parkinsonism, specifically in the metabolism of MPTP, which selectively targets dopaminergic neurons [142,143]. Within astrocytes, MAO-B converts MPTP into its toxic metabolite MPP+, which enters dopaminergic neurons via the dopamine transporter, leading to mitochondrial dysfunction and cell death [143-145]. MAO-B inhibitors such as selegiline mitigate MPTP-induced neurotoxicity, suggesting neuroprotective mechanisms beyond DA metabolism, potentially involving mitochondrial and glial function [114,118].

These findings highlight the complex role of MAO-B in PD, extending beyond DA metabolism to astrocytic regulation and neuroinflammatory pathways.

MAO-B in PD: Astrocytic GABA regulation and oxidative stress

MAO-B has long been recognized for its role in dopamine metabolism and the activation of the neurotoxin MPTP into its toxic form, MPP+, contributing to PD pathogenesis. However, recent studies have expanded its functional scope beyond DA degradation, highlighting its involvement in astrocytic GABA synthesis and H2O2 production—both of which significantly contribute to neurodegeneration [128,146-148].

Under physiological conditions, MAO-B regulates astrocytic GABA synthesis, particularly in the cerebellum and striatum [17,149]. It catalyzes the conversion of putrescine into GABA, which is subsequently released tonically through the Ca²+-activated anion channel Bestrophin-1 (Best1) [9,17]. This extrasynaptic GABA exerts inhibitory effects on neighboring neurons, with a region-specific distribution favoring the striatum and cerebellum [150,151]. Unlike MAO-B, MAO-A does not participate in astrocytic GABA-mediated inhibition in the striatum, reinforcing MAO-B’s specialized role in modulating neuronal excitability [2,152].

In PD, reactive astrogliosis leads to increased MAO-B expression in the substantia nigra pars compacta (SNpc), driving excessive astrocytic GABA synthesis and abnormal tonic inhibition of dopaminergic neurons [9,126,153]. This pathological inhibition suppresses neuronal activity and downregulates tyrosine hydroxylase (TH), a key enzyme in DA synthesis [129,154]. While TH suppression is reversible in viable neurons, prolonged inhibition exacerbates DA depletion and contributes to PD-associated motor deficits [10,77,155].

Beyond GABA synthesis, MAO-B also catalyzes the degradation of N-acetylputrescine, generating H2O2 as a metabolic byproduct [1,9]. Excessive H2O2 in PD exacerbates neurodegeneration by promoting oxidative stress, enhancing astrocytic reactivity, and accelerating dopaminergic neuron loss [34,90,156]. Experimental evidence from MPTP mouse models suggests that scavenging H2O2 can mitigate neurodegeneration, further implicating MAO-B-mediated oxidative stress in PD pathology [79,143,156].

The emerging understanding of MAO-B’s role in astrocytic GABA regulation and oxidative stress has redefined its contribution to PD, shifting the focus beyond DA metabolism. These findings establish MAO-B as a key player in pathological astrocytic signaling and highlight its potential as a therapeutic target in PD.

The Role of MAO-B in GABA Regulation and AD Pathogenesis

MAO-B has emerged as a crucial factor in the dysregulation of GABAergic signaling in AD [70,71]. As an enzyme responsible for the oxidative deamination of neurotransmitters, such as dopamine and serotonin, MAO-B produces ROS as byproducts, contributing significantly to oxidative stress in the brain. Under normal conditions, MAO-B is present at low levels in the brain, but its expression is markedly upregulated in reactive astrocytes during neurodegenerative diseases, including AD [71,98,140,157]. The increased activity of MAO-B in these glial cells has profound implications for neurotransmitter metabolism and neuronal health, particularly in the context of GABA regulation (Table 3) [2,118,158].

Table 3. Impact of MAO-B Activity on Astrocytic GABA Regulation in Alzheimer's Disease Pathogenesis.

Key Element

Description

Mechanisms Involved

Implications for AD Pathophysiology

Therapeutic Considerations

References

MAO-B Activity in Astrocytes

MAO-B is overexpressed in reactive astrocytes in AD, leading to oxidative stress and disrupted GABA metabolism.

MAO-B deaminates neurotransmitters like dopamine, generating reactive oxygen species (ROS) as byproducts.

Elevated MAO-B activity exacerbates oxidative stress, impairing GABA metabolism and synaptic function.

Inhibiting MAO-B could reduce oxidative stress and restore GABAergic balance in AD.

[12,44,70]

MAO-B and GABA Transporters

MAO-B activity impairs GABA transporters (GATs) in astrocytes, reducing the reuptake of GABA from the synaptic cleft.

MAO-B-induced ROS damage compromises GAT-1 function, leading to abnormal accumulation of GABA.

Dysregulated GABA uptake contributes to excessive extracellular GABA, worsening excitatory-inhibitory imbalance.

Restoring GAT function or reducing MAO-B activity may help re-establish proper GABAergic tone.

[12,159]

MAO-B and GABA Synthesis

Increased MAO-B activity in astrocytes reduces the synthesis of GABA by disrupting the enzymatic processes.

MAO-B-induced oxidative stress damages enzymes involved in GABA synthesis, such as GAD.

Impaired GABA synthesis contributes to reduced inhibitory signaling, promoting neuronal hyperactivity.

Targeting MAO-B to alleviate oxidative damage could enhance GABA synthesis and improve inhibition.

[2,17,160]

MAO-B and GABA Receptor Function

MAO-B overexpression in astrocytes affects GABA receptor expression and function, impairing synaptic plasticity.

Altered GABA-A and GABA-B receptor function due to ROS accumulation from MAO-B activity.

Dysfunctional GABA receptors exacerbate synaptic dysfunction, impairing plasticity and cognitive processes.

Pharmacologically targeting GABA-A and GABA-B receptors could mitigate synaptic dysfunction in AD.

[12,74]

MAO-B and Astrocyte-Neuron Interactions

MAO-B influences astrocytic support for neurons, affecting synaptic GABAergic function in AD.

Reactive astrocytes with high MAO-B activity affect neuronal excitability and GABAergic signaling.

MAO-B-induced damage to astrocytes disrupts their ability to modulate neuronal activity, exacerbating neurodegeneration.

Modulating MAO-B could protect astrocyte-neuron interactions and maintain GABAergic signaling.

[12,17,161]

MAO-B and Neuroinflammation

Elevated MAO-B activity enhances pro-inflammatory cytokine release from astrocytes, exacerbating AD pathology.

MAO-B activity in astrocytes stimulates inflammatory pathways, contributing to neuroinflammation.

Neuroinflammation worsens GABAergic dysfunction, leading to synaptic loss and cognitive decline.

Inhibiting MAO-B could reduce neuroinflammation, improving GABAergic function and slowing AD.

[18,70,162]

MAO-B and Excitotoxicity

MAO-B-induced oxidative stress promotes neuronal hyperexcitability by impairing GABAergic inhibition.

The ROS produced by MAO-B activity in astrocytes leads to neuronal overactivation and excitotoxicity.

Impaired GABAergic inhibition due to MAO-B activity increases the risk of excitotoxicity and neuronal damage.

MAO-B inhibition could protect neurons from excitotoxic damage by restoring GABAergic function.

[71,118]

Therapeutic Potential: MAO-B Inhibition

MAO-B inhibitors, such as selegiline, have shown potential in reducing oxidative stress and restoring GABAergic balance.

MAO-B inhibitors decrease ROS production, protecting GABA transporters and improving synaptic function.

Inhibiting MAO-B may improve cognitive function, reduce neuroinflammation, and alleviate oxidative damage.

Selegiline and similar MAO-B inhibitors could provide therapeutic benefit in AD by restoring GABA signaling.

[71,114,163]

Alternative Therapeutic Strategies

Targeting GABA-A or GABA-B receptors on astrocytes may restore GABAergic signaling disrupted by MAO-B.

Modulating GABA receptor function on astrocytes can help re-establish inhibitory control.

Restoring proper GABAergic tone could protect against cognitive decline and neurodegeneration in AD.

Developing drugs to modulate astrocytic GABA receptors could complement MAO-B inhibition for AD therapy.

[39,160]


In AD, the overexpression of MAO-B in reactive astrocytes exacerbates oxidative stress, which disrupts GABAergic signaling [71,74,164]. GABA is essential for regulating neuronal excitability and maintaining synaptic balance [165,166]. Astrocytes are responsible for maintaining GABA homeostasis through the uptake and clearance of excess GABA from the synaptic cleft via GABA transporters (GATs) [39,148]. However, elevated MAO-B activity generates ROS that can damage these transporters, impairing their ability to effectively clear GABA from the synaptic space [167,168]. This impairment leads to the accumulation of extracellular GABA, which disrupts the balance between excitatory and inhibitory signaling in the brain, resulting in neuronal hyperexcitability and contributing to the cognitive deficits observed in AD (Table 3) [169-171].

The role of MAO-B in GABA metabolism is not limited to its effect on GABA transporters. The enzyme's activity also affects GABA synthesis. ROS generated by MAO-B can inhibit key enzymes involved in GABA production, thereby reducing the availability of GABA in the brain [71,161,169]. This reduction further exacerbates GABAergic dysfunction and enhances neuronal excitability, ultimately accelerating neurodegenerative processes in AD [17]. Additionally, MAO-B-mediated oxidative stress can interfere with the functionality of GABA receptors, such as GABA-A and GABA-B receptors, which are involved in astrocyte responses to GABAergic activity (Table 3) [12,39,172]. Disruption of GABA receptor signaling impairs communication between astrocytes and neurons, compounding the GABAergic dysregulation seen in AD and contributing to the neurodegenerative cascade [74,171,173,174].

Furthermore, MAO-B-induced oxidative stress may have a synergistic effect when coupled with other pathophysiological processes in AD, such as the accumulation of amyloid-β (Aβ) plaques [175,176]. Aβ plaques are known to trigger inflammatory responses in astrocytes, leading to reactive gliosis and further upregulation of MAO-B [70,74,150]. This creates a vicious cycle in which MAO-B activity promotes oxidative damage, which in turn exacerbates the neuroinflammation and GABAergic dysfunction characteristic of AD (Table 3) [12,74]. As oxidative stress accumulates contributing to the progression of cognitive decline and neurodegeneration.

Given the central role of MAO-B in GABAergic dysfunction and oxidative stress, it has become an attractive therapeutic target for AD. Inhibiting MAO-B activity could potentially reduce ROS production, restore GABA homeostasis, and alleviate the neuronal excitability that drives neurodegeneration. Several MAO-B inhibitors are already used in the treatment of PD, and their application in AD holds promise for modulating oxidative stress and improving GABAergic signaling (Table 3). Furthermore, understanding the molecular mechanisms that govern MAO-B expression and activity in astrocytes could provide new insights into the pathogenesis of AD and lead to the development of more targeted and effective treatments.

Role of MAO-B in Astrocyte Reactivity and Glial Scar Formation Following CNS Injury

MAO-B plays a critical role in astrocyte reactivity and glial scar formation, key processes in the brain's response to injury [82,177]. Reactive astrocytes undergo substantial morphological and biochemical changes, such as hypertrophy and metabolic reprogramming, which are necessary for the formation of glial scars that help isolate the injured area [178,179]. One of the central mediators in this process is H2O2, a by-product of various enzymatic reactions, which is thought to be instrumental in initiating astrocytic hypertrophy and scar formation [152,180]. MAO-B contributes to H2O2 production, which is associated with astrocyte dysfunction and the progression of glial scar formation [153,177].

Astrocyte reactivity is tightly controlled by specific enzymatic pathways, with the MAO-B-H2O2 signaling axis playing a pivotal role in driving astrocytic hypertrophy and proliferation [90,148,181]. Increased MAO-B activity leads to higher H2O2 production, which in turn promotes the transformation of astrocytes into a hypertrophic, scar-forming phenotype [34,153]. This process is further influenced by interactions between reactive astrocytes and other cell types, such as microglia, which collectively contribute to the inflammatory response following CNS injury. Inhibiting MAO-B activity has shown potential in reducing astrocyte reactivity and glial scar formation, highlighting the therapeutic potential of targeting this pathway in neurodegenerative diseases and CNS injuries [82,150,153].

While glial scars formed by reactive astrocytes serve a protective role by isolating damaged regions and preventing further neuronal injury, they also hinder axonal regeneration, complicating recovery. Reactive astrocytes exhibit varying degrees of reactivity, from mild hypertrophy to more severe hypertrophy and proliferation, likely due to the activation of different metabolic pathways. Balancing the reduction of scar formation while maintaining the protective functions of astrocytes is a significant challenge. Modulating MAO-B activity offers a potential strategy to shift astrocytes toward a less reactive state, thereby mitigating the adverse effects of glial scar formation while preserving their critical neuroprotective roles in the injury response.

Monoamine Oxidase B in Astrocytic GABA Synthesis and Cancer Biology

MAO-B is recognized for its role in neurotransmitter metabolism within the central nervous system. However, recent studies have expanded its significance, revealing it as a crucial player in various cancer-related processes [182]. In addition to its established neurobiological functions, MAO-B has been implicated in cancer progression, particularly through its regulation of oxidative stress, modulation of the tumor microenvironment (TME), and influence on cellular dynamics [183]. Notably, MAO-B's involvement in astrocytic GABA synthesis adds further complexity to neuro-oncological contexts, such as gliomas, where it may mediate interactions between neurons and glial cells, ultimately affecting tumor behavior.

Mechanisms of MAO-B in cancer biology

One of the primary mechanisms through which MAO-B contributes to cancer progression is its regulation of ROS. MAO-B catalyzes the breakdown of biogenic amines, including dopamine, generating hydrogen peroxide as a byproduct [107]. This process leads to the accumulation of ROS, which induces oxidative stress, an established factor in cancer development [184]. The accumulation of ROS can result in DNA damage, activation of inflammatory responses, and disruption of cellular signaling, all of which promote genomic instability and create a microenvironment conducive to malignant transformation [184]. By enhancing oxidative stress, MAO-B thus facilitates early-stage cancer development and tumor progression.

In addition to ROS regulation, MAO-B overexpression is commonly observed in various cancers, where it influences the TME [41]. MAO-B alters metabolic reprogramming [2,185] by shifting the balance between aerobic and anaerobic metabolism—processes that are essential for cancer cell survival and proliferation. This metabolic shift contributes to therapeutic resistance. Moreover, the inflammatory environment generated by MAO-B activity further complicates the TME [103,186], promoting immune evasion and enhancing metastatic potential. Thus, MAO-B becomes integral to the maintenance of the malignant state, supporting both local tumor growth and metastatic spread.

MAO-B also affects cancer cell motility and invasion [187], which are crucial factors in metastasis. By modulating key signaling pathways related to cell adhesion, migration, and extracellular matrix remodeling, MAO-B enhances the ability of cancer cells to invade surrounding tissues and spread to distant organs. This metastatic potential underscore MAO-B's critical role not only in local tumor growth but also in cancer cell dissemination [182].

MAO-B in astrocytic GABA synthesis and cancer

Beyond its role in cancer biology, MAO-B plays a critical role in neurotransmitter metabolism, particularly in the synthesis of GABA [9]. Within astrocytes, MAO-B is involved in the catabolism of GABA precursors, such as the conversion of glutamate to glutamine [38,188], which is subsequently utilized in GABA synthesis. This process is essential for maintaining the balance of neurotransmitter pools in the brain, particularly in regions responsible for regulating neuronal excitability and synaptic plasticity, such as the cortex and hippocampus.

The involvement of MAO-B in astrocytic GABA synthesis takes on particular significance in brain tumors, especially gliomas, where the balance between excitatory and inhibitory signals is often disrupted [39]. In these tumors, altered astrocytic function, including changes in GABAergic signaling [189,190], may be influenced by MAO-B activity. Dysregulation of GABAergic signaling in gliomas can significantly affect tumor cell behavior, including proliferation, survival, and communication between tumor cells and surrounding glial cells [191]. This disruption may contribute to abnormal TME, which supports tumor growth and resistance to therapies.

Furthermore, astrocytic GABA synthesis is essential for proper neuronal-glial communication [39]. In gliomas, this communication is often impaired, and the altered GABAergic signaling can affect the functional interactions between glial cells and tumor cells. Such dysregulation complicates the tumor's response to treatment, further emphasizing the complex role of MAO-B in regulating astrocytic GABA synthesis within brain tumors.

Discussion and Concluding Remarks

MAO-B plays a multifaceted role in neurological health and disease, impacting both neurotransmitter regulation and neuroinflammatory processes. As highlighted in this article, MAO-B is essential for the oxidative deamination of key monoamines such as dopamine, noradrenaline, and serotonin, but it also significantly influences astrocytic modulation of neurotransmitter signaling, particularly GABA. The article explores the dual nature of MAO-B activity: while it is vital for maintaining normal brain function, its dysregulation can lead to neuroinflammation, oxidative stress, and neuronal damage, contributing to the pathophysiology of a range of neurological disorders, including AD, PD, HD, and ALS.

In neurodegenerative diseases such as AD and PD, heightened MAO-B activity in astrocytes leads to the generation of ROS, including hydrogen peroxide, which exacerbates oxidative stress and GABAergic dysfunction. This disruption of neurotransmitter balance accelerates neuronal loss and impairs neuronal recovery. The article also discusses how MAO-B-induced glial reactivity contributes to the formation of glial scars, which, while protective, hinder axonal regeneration and recovery. Targeting these scars offers a promising therapeutic intervention.

Future research should focus on uncovering the mechanisms through which MAO-B modulates neuroinflammation and neurotransmitter imbalances, particularly within reactive astrocytes. Examining how MAO-B activity affects neuronal function in specific brain regions, such as the hypothalamus and cerebellum, could provide critical insights into disease-specific pathophysiology and potential therapeutic targets. Additionally, the therapeutic potential of MAO-B inhibitors, which have shown promise in alleviating symptoms and slowing disease progression in conditions like PD, should be further explored in clinical trials to determine their efficacy in a broader range of neurodegenerative and neuroinflammatory diseases.

Identify MAO-B as a potential enzyme responsible for synthesizing GABA within LV NSCs, which regulate their homeostatic proliferation through neural circuit activity. This discovery could provide deeper insights into glioblastoma and its regulation by neural circuits, given its presumed origin in LV NSCs. Furthermore, investigating whether glioblastoma synthesizes GABA, whether this synthesis occurs through MAO-B, and how GABA contributes to glioblastoma's sustained growth and progression warrants further exploration.

The enzymatic activity of MAO-B in tumors influences aberrant signaling pathways, contributing to tumor growth, treatment resistance, and metastasis. These findings suggest that MAO-B inhibitors may serve as potential therapeutic agents in cancer treatment. Early studies have shown that inhibiting MAO-B suppresses cancer cell proliferation across various tumor types, highlighting a promising approach to targeting oxidative stress and tumor dynamics driven by MAO-B. The dual role of MAO-B in both cancer biology and neurotransmitter metabolism offers new insights into the intricate relationship between the nervous system and cancer. Additionally, understanding the impact of MAO-B on astrocytic GABA synthesis in brain tumors provides valuable perspectives on the molecular mechanisms governing tumor behavior and neuron-glial interactions. Modulating MAO-B’s enzymatic activity may help address both oxidative stress and neurotransmitter imbalances in tumors, paving the way for innovative therapeutic strategies in neuro-oncology.

One limitation of the current body of research is the complexity of MAO-B’s dual role in both neurotransmitter regulation and neuroinflammation. The effects of MAO-B upregulation in reactive astrocytes are context-dependent and may vary across different neurological conditions, complicating our understanding of its precise role at various stages of disease progression. Furthermore, although MAO-B inhibitors have demonstrated therapeutic potential, their inconsistent efficacy underscores the need for a more nuanced approach to targeting MAO-B without disrupting essential brain functions.

To address these gaps, future studies should aim to clarify the temporal dynamics of MAO-B activity in neurodegenerative diseases, assessing whether modulation in the early stages offers protective effects or if sustained inhibition could prevent progression in later stages. Longitudinal studies, coupled with advanced imaging technologies, may provide valuable insights into the impact of MAO-B inhibition on neuroinflammation, synaptic plasticity, and astrocyte function. Moreover, expanding research into the effects of MAO-B modulation in other conditions, such as metabolic disorders and traumatic brain injury, could broaden the therapeutic potential of targeting this enzyme.

MAO-B is a pivotal enzyme with profound implications for both brain health and the pathogenesis of neurological diseases. While its upregulation in reactive astrocytes contributes to neurodegeneration by exacerbating oxidative stress and disrupting GABAergic signaling, MAO-B also represents a promising therapeutic target for neurodegenerative disorders. This article highlights the potential of MAO-B inhibitors to slow disease progression and restore neurotransmitter homeostasis. However, it emphasizes the necessity of a more refined understanding of MAO-B’s complex and context-dependent roles in brain function. Future research should prioritize elucidating the mechanisms that govern MAO-B regulation across various neurological conditions, with the aim of optimizing its therapeutic targeting in neurodegenerative and metabolic diseases.

Declarations

Acknowledgments

N/A.

Author contributions

M.M.N. contributed to the design and writing of the main manuscript text.

Conflicts of interest

The author declares no conflict of interest.

Ethical approval

Not applicable.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of Data and Materials

Not applicable.

Funding

This research received no external funding.

References

1. Bortolato M, Chen K, Shih JC. Monoamine oxidase inactivation: from pathophysiology to therapeutics. Adv Drug Deliv Rev. 2008 Oct-Nov;60(13-14):1527-33.

2. Cho HU, Kim S, Sim J, Yang S, An H, Nam MH, et al. Redefining differential roles of MAO-A in dopamine degradation and MAO-B in tonic GABA synthesis. Exp Mol Med. 2021 Jul;53(7):1148-58.

3. Mentis AA, Dardiotis E, Katsouni E, Chrousos GP. From warrior genes to translational solutions: novel insights into monoamine oxidases (MAOs) and aggression. Transl Psychiatry. 2021 Feb 18;11(1):130.

4. Singh C, Bortolato M, Bali N, Godar SC, Scott AL, Chen K, et al. Cognitive abnormalities and hippocampal alterations in monoamine oxidase A and B knockout mice. Proc Natl Acad Sci U S A. 2013 Jul 30;110(31):12816-21.

5. Uzbekov MG. Monoamine Oxidase as a Potential Biomarker of the Efficacy of Treatment of Mental Disorders. Biochemistry (Mosc). 2021 Jun;86(6):773-83.

6. Kaludercic N, Carpi A, Menabò R, Di Lisa F, Paolocci N. Monoamine oxidases (MAO) in the pathogenesis of heart failure and ischemia/reperfusion injury. Biochim Biophys Acta. 2011 Jul;1813(7):1323-32.

7. Bergström M, Westerberg G, Långström B. 11C-harmine as a tracer for monoamine oxidase A (MAO-A): in vitro and in vivo studies. Nucl Med Biol. 1997 May;24(4):287-93.

8. Obata Y, Kubota-Sakashita M, Kasahara T, Mizuno M, Nemoto T, Kato T. Phenethylamine is a substrate of monoamine oxidase B in the paraventricular thalamic nucleus. Sci Rep. 2022 Jan 7;12(1):17.

9. Nam MH, Sa M, Ju YH, Park MG, Lee CJ. Revisiting the Role of Astrocytic MAOB in Parkinson's Disease. Int J Mol Sci. 2022 Apr 18;23(8):4453.

10. Masato A, Plotegher N, Boassa D, Bubacco L. Impaired dopamine metabolism in Parkinson's disease pathogenesis. Mol Neurodegener. 2019 Aug 20;14(1):35.

11. Graves SM, Xie Z, Stout KA, Zampese E, Burbulla LF, Shih JC, et al. Dopamine metabolism by a monoamine oxidase mitochondrial shuttle activates the electron transport chain. Nat Neurosci. 2020 Jan;23(1):15-20.

12. Jo S, Yarishkin O, Hwang YJ, Chun YE, Park M, Woo DH, et al. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease. Nat Med. 2014 Aug;20(8):886-96.

13. Kilb W, Kirischuk S. GABA Release from Astrocytes in Health and Disease. Int J Mol Sci. 2022 Dec 13;23(24):15859.

14. Lee N, Sa M, Hong YR, Lee CJ, Koo J. Fatty Acid Increases cAMP-dependent Lactate and MAO-B-dependent GABA Production in Mouse Astrocytes by Activating a Gαs Protein-coupled Receptor. Exp Neurobiol. 2018 Oct;27(5):365-76.

15. Ness N, Díaz-Clavero S, Hoekstra MMB, Brancaccio M. Rhythmic astrocytic GABA production synchronizes neuronal circadian timekeeping in the suprachiasmatic nucleus. EMBO J. 2025 Jan;44(2):356-81.

16. Bhalla M, Joo J, Kim D, Shin JI, Park YM, Ju YH, et al. SIRT2 and ALDH1A1 as critical enzymes for astrocytic GABA production in Alzheimer's disease. Mol Neurodegener. 2025 Jan 15;20(1):6.

17. Yoon BE, Woo J, Chun YE, Chun H, Jo S, Bae JY, et al. Glial GABA, synthesized by monoamine oxidase B, mediates tonic inhibition. J Physiol. 2014 Nov 15;592(22):4951-68.

18. Carter SF, Schöll M, Almkvist O, Wall A, Engler H, Långström B, et al. Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG. J Nucl Med. 2012 Jan;53(1):37-46.

19. Schedin-Weiss S, Inoue M, Hromadkova L, Teranishi Y, Yamamoto NG, Wiehager B, et al. Monoamine oxidase B is elevated in Alzheimer disease neurons, is associated with γ-secretase and regulates neuronal amyloid β-peptide levels. Alzheimers Res Ther. 2017 Aug 1;9(1):57.

20. Matveychuk D, MacKenzie EM, Kumpula D, Song MS, Holt A, Kar S, et al. Overview of the Neuroprotective Effects of the MAO-Inhibiting Antidepressant Phenelzine. Cell Mol Neurobiol. 2022 Jan;42(1):225-42.

21. Arcone R, D'Errico A, Nasso R, Rullo R, Poli A, Di Donato P, et al. Inhibition of Enzymes Involved in Neurodegenerative Disorders and Aβ1-40 Aggregation by Citrus limon Peel Polyphenol Extract. Molecules. 2023 Aug 30;28(17):6332.

22. Naffaa, M., and Yin, H. . Lateral Ventricular Neural Stem Cells (LV NSCs) Regulate Neural Circuitry Activity Through GABAergic Signaling. Preprints 2025, 2025011266.

23. Naffaa MM, Khan RR, Kuo CT, Yin HH. Cortical regulation of neurogenesis and cell proliferation in the ventral subventricular zone. Cell Rep. 2023 Jul 25;42(7):112783.

24. Naffaa MM, Yin HH. Lateral Ventricular Neural Stem Cells Provide Negative Feedback to Circuit Activation Through GABAergic Signaling. Cells. 2025 Mar 13;14(6):426.

25. Wang CC, Billett E, Borchert A, Kuhn H, Ufer C. Monoamine oxidases in development. Cell Mol Life Sci. 2013 Feb;70(4):599-630.

26. Bortolato M, Shih JC. Behavioral outcomes of monoamine oxidase deficiency: preclinical and clinical evidence. Int Rev Neurobiol. 2011;100:13-42.

27. Yeung AWK, Georgieva MG, Atanasov AG, Tzvetkov NT. Monoamine Oxidases (MAOs) as Privileged Molecular Targets in Neuroscience: Research Literature Analysis. Front Mol Neurosci. 2019 May 29;12:143.

28. O'Carroll AM, Fowler CJ, Phillips JP, Tobbia I, Tipton KF. The deamination of dopamine by human brain monoamine oxidase. Specificity for the two enzyme forms in seven brain regions. Naunyn Schmiedebergs Arch Pharmacol. 1983 Apr;322(3):198-202.

29. Tong J, Meyer JH, Furukawa Y, Boileau I, Chang LJ, Wilson AA, et al. Distribution of monoamine oxidase proteins in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab. 2013 Jun;33(6):863-71.

30. Vitalis T, Fouquet C, Alvarez C, Seif I, Price D, Gaspar P, et al. Developmental expression of monoamine oxidases A and B in the central and peripheral nervous systems of the mouse. J Comp Neurol. 2002 Jan 21;442(4):331-47.

31. Luque JM, Kwan SW, Abell CW, Da Prada M, Richards JG. Cellular expression of mRNAs encoding monoamine oxidases A and B in the rat central nervous system. J Comp Neurol. 1995 Dec 25;363(4):665-80.

32. Levitt P, Pintar JE, Breakefield XO. Immunocytochemical demonstration of monoamine oxidase B in brain astrocytes and serotonergic neurons. Proc Natl Acad Sci U S A. 1982 Oct;79(20):6385-9.

33. Gill T, Watling SE, Richardson JD, McCluskey T, Tong J, Meyer JH, et al. Imaging of astrocytes in posttraumatic stress disorder: A PET study with the monoamine oxidase B radioligand [11C] SL25. 1188. Eur Neuropsychopharmacol. 2022 Jan 1;54:54-61.

34. Mallajosyula JK, Kaur D, Chinta SJ, Rajagopalan S, Rane A, Nicholls DG, et al. MAO-B elevation in mouse brain astrocytes results in Parkinson's pathology. PLoS One. 2008 Feb 20;3(2):e1616.

35. Saura J, Andrés N, Andrade C, Ojuel J, Eriksson K, Mahy N. Biphasic and region-specific MAO-B response to aging in normal human brain. Neurobiol Aging. 1997 Sep-Oct;18(5):497-507.

36. Sharvin BL, Aburto MR, Cryan JF. Decoding the neurocircuitry of gut feelings: Region-specific microbiome-mediated brain alterations. Neurobiol Dis. 2023 Apr;179:106033.

37. Nugraha RYB, Jeelani G, Nozaki T. Physiological roles and metabolism of γ-aminobutyric acid (GABA) in parasitic protozoa. Trends Parasitol. 2022 Jun;38(6):462-77.

38. Andersen JV, Westi EW, Jakobsen E, Urruticoechea N, Borges K, Aldana BI. Astrocyte metabolism of the medium-chain fatty acids octanoic acid and decanoic acid promotes GABA synthesis in neurons via elevated glutamine supply. Mol Brain. 2021 Sep 3;14(1):132.

39. Liu J, Feng X, Wang Y, Xia X, Zheng JC. Astrocytes: GABAceptive and GABAergic Cells in the Brain. Front Cell Neurosci. 2022 Jun 10;16:892497.

40. Sharif A, Prevot V. Astrogenesis in the hypothalamus: A life-long process contributing to the development and plasticity of neuroendocrine networks. Front Neuroendocrinol. 2024 Oct;75:101154.

41. Sharpe MA, Baskin DS. Monoamine oxidase B levels are highly expressed in human gliomas and are correlated with the expression of HiF-1α and with transcription factors Sp1 and Sp3. Oncotarget. 2016 Jan 19;7(3):3379-93.

42. Sheng WS, Hu S, Feng A, Rock RB. Reactive oxygen species from human astrocytes induced functional impairment and oxidative damage. Neurochem Res. 2013 Oct;38(10):2148-59.

43. Calì C, Cantando I, Veloz Castillo MF, Gonzalez L, Bezzi P. Metabolic Reprogramming of Astrocytes in Pathological Conditions: Implications for Neurodegenerative Diseases. Int J Mol Sci. 2024 Aug 16;25(16):8922.

44. Park JH, Ju YH, Choi JW, Song HJ, Jang BK, Woo J, et al. Newly developed reversible MAO-B inhibitor circumvents the shortcomings of irreversible inhibitors in Alzheimer's disease. Sci Adv. 2019 Mar 20;5(3):eaav0316.

45. Hussaini SMQ, Jang MH. New Roles for Old Glue: Astrocyte Function in Synaptic Plasticity and Neurological Disorders. Int Neurourol J. 2018 Oct;22(Suppl 3):S106-114.

46. Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, et al. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci. 2023 Jan 19;16:1037641.

47. Sahlender DA, Savtchouk I, Volterra A. What do we know about gliotransmitter release from astrocytes? Philos Trans R Soc Lond B Biol Sci. 2014 Oct 19;369(1654):20130592.

48. Murat CB, García-Cáceres C. Astrocyte Gliotransmission in the Regulation of Systemic Metabolism. Metabolites. 2021 Oct 26;11(11):732.

49. Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. Front Netw Physiol. 2023 Jun 1;3:1205544.

50. Untiet V. Astrocytic chloride regulates brain function in health and disease. Cell Calcium. 2024 Mar;118:102855.

51. Lee S, Yoon BE, Berglund K, Oh SJ, Park H, Shin HS, et al. Channel-mediated tonic GABA release from glia. Science. 2010 Nov 5;330(6005):790-6.

52. Liu X, Ying J, Wang X, Zheng Q, Zhao T, Yoon S, et al. Astrocytes in Neural Circuits: Key Factors in Synaptic Regulation and Potential Targets for Neurodevelopmental Disorders. Front Mol Neurosci. 2021 Sep 29;14:729273.

53. Woo J, Min JO, Kang DS, Kim YS, Jung GH, Park HJ, et al. Control of motor coordination by astrocytic tonic GABA release through modulation of excitation/inhibition balance in cerebellum. Proc Natl Acad Sci U S A. 2018 May 8;115(19):5004-9.

54. Ishibashi M, Egawa K, Fukuda A. Diverse Actions of Astrocytes in GABAergic Signaling. Int J Mol Sci. 2019 Jun 18;20(12):2964.

55. Zimmer TS, Orr AL, Orr AG. Astrocytes in selective vulnerability to neurodegenerative disease. Trends Neurosci. 2024 Apr;47(4):289-302.

56. Ben Haim L, Carrillo-de Sauvage MA, Ceyzériat K, Escartin C. Elusive roles for reactive astrocytes in neurodegenerative diseases. Front Cell Neurosci. 2015 Aug 3;9:278.

57. Teleanu RI, Niculescu AG, Roza E, Vladâcenco O, Grumezescu AM, Teleanu DM. Neurotransmitters-Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Int J Mol Sci. 2022 May 25;23(11):5954.

58. Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, et al. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun. 2024 Mar 12;15(1):2219.

59. Alharbi B, Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Alharbi H, Alexiou A, et al. Role of GABA pathway in motor and non-motor symptoms in Parkinson's disease: a bidirectional circuit. Eur J Med Res. 2024 Mar 27;29(1):205.

60. O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci. 2020 Jul 23;14:187.

61. Choudhury GR, Ding S. Reactive astrocytes and therapeutic potential in focal ischemic stroke. Neurobiol Dis. 2016 Jan;85:234-44.

62. Obara-Michlewska M. The contribution of astrocytes to obesity-associated metabolic disturbances. J Biomed Res. 2022 Aug 28;36(5):299-311.

63. Naveed M, Smedlund K, Zhou QG, Cai W, Hill JW. Astrocyte involvement in metabolic regulation and disease. Trends Endocrinol Metab. 2025 Mar;36(3):219-34.

64. Perucca E, White HS, Bialer M. New GABA-Targeting Therapies for the Treatment of Seizures and Epilepsy: II. Treatments in Clinical Development. CNS Drugs. 2023 Sep;37(9):781-95.

65. Burman RJ, Selfe JS, Lee JH, van den Berg M, Calin A, Codadu NK, et al. Excitatory GABAergic signalling is associated with benzodiazepine resistance in status epilepticus. Brain. 2019 Nov 1;142(11):3482-501.

66. Subramaniam SR, Chesselet MF. Mitochondrial dysfunction and oxidative stress in Parkinson's disease. Prog Neurobiol. 2013 Jul-Aug;106-107:17-32.

67. Niedzielska E, Smaga I, Gawlik M, Moniczewski A, Stankowicz P, Pera J, et al. Oxidative Stress in Neurodegenerative Diseases. Mol Neurobiol. 2016 Aug;53(6):4094-125.

68. Sen MK, Mahns DA, Coorssen JR, Shortland PJ. The roles of microglia and astrocytes in phagocytosis and myelination: Insights from the cuprizone model of multiple sclerosis. Glia. 2022 Jul;70(7):1215-50.

69. Rajda C, Pukoli D, Bende Z, Majláth Z, Vécsei L. Excitotoxins, Mitochondrial and Redox Disturbances in Multiple Sclerosis. Int J Mol Sci. 2017 Feb 8;18(2):353.

70. Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol. 2024 Apr 3;147(1):66.

71. Behl T, Kaur D, Sehgal A, Singh S, Sharma N, Zengin G, et al. Role of Monoamine Oxidase Activity in Alzheimer's Disease: An Insight into the Therapeutic Potential of Inhibitors. Molecules. 2021 Jun 18;26(12):3724.

72. Dröge W, Schipper HM. Oxidative stress and aberrant signaling in aging and cognitive decline. Aging Cell. 2007 Jun;6(3):361-70.

73. Labarta-Bajo L, Allen NJ. Astrocytes in aging. Neuron. 2025 Jan 8;113(1):109-26.

74. González-Reyes RE, Nava-Mesa MO, Vargas-Sánchez K, Ariza-Salamanca D, Mora-Muñoz L. Involvement of Astrocytes in Alzheimer's Disease from a Neuroinflammatory and Oxidative Stress Perspective. Front Mol Neurosci. 2017 Dec 19;10:427.

75. Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev. 2021 Jul;68:101335.

76. Rodriguez-Vieitez E, Nordberg A. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol. 2018;1750:231-251.

77. Chakrabarti S, Bisaglia M. Oxidative Stress and Neuroinflammation in Parkinson's Disease: The Role of Dopamine Oxidation Products. Antioxidants (Basel). 2023 Apr 18;12(4):955.

78. Guo JD, Zhao X, Li Y, Li GR, Liu XL. Damage to dopaminergic neurons by oxidative stress in Parkinson's disease (Review). Int J Mol Med. 2018 Apr;41(4):1817-25.

79. Imbriani P, Martella G, Bonsi P, Pisani A. Oxidative stress and synaptic dysfunction in rodent models of Parkinson's disease. Neurobiol Dis. 2022 Oct 15;173:105851.

80. Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol. 2019 Feb 1;316(2):C135-53.

81. Collyer E, Blanco-Suarez E. Astrocytes in stroke-induced neurodegeneration: a timeline. Front Mol Med. 2023 Sep 7;3:1240862.

82. Cieri MB, Ramos AJ. Astrocytes, reactive astrogliosis, and glial scar formation in traumatic brain injury. Neural Regen Res. 2025 Apr 1;20(4):973-89.

83. Zheng RZ, Lee KY, Qi ZX, Wang Z, Xu ZY, Wu XH, et al. Neuroinflammation Following Traumatic Brain Injury: Take It Seriously or Not. Front Immunol. 2022 Mar 22;13:855701.

84. Correale J, Farez MF. The Role of Astrocytes in Multiple Sclerosis Progression. Front Neurol. 2015 Aug 18;6:180.

85. Colón Ortiz C, Eroglu C. Astrocyte signaling and interactions in Multiple Sclerosis. Curr Opin Cell Biol. 2024 Feb;86:102307.

86. Ellen O, Ye S, Nheu D, Dass M, Pagnin M, Ozturk E, et al. The Heterogeneous Multiple Sclerosis Lesion: How Can We Assess and Modify a Degenerating Lesion? Int J Mol Sci. 2023 Jul 5;24(13):11112.

87. Harada R, Furumoto S, Kudo Y, Yanai K, Villemagne VL, Okamura N. Imaging of Reactive Astrogliosis by Positron Emission Tomography. Front Neurosci. 2022 Feb 8;16:807435.

88. Johansson A, Engler H, Blomquist G, Scott B, Wall A, Aquilonius SM, Långström B, Askmark H. Evidence for astrocytosis in ALS demonstrated by [11C](L)-deprenyl-D2 PET. J Neurol Sci. 2007 Apr 15;255(1-2):17-22.

89. Richards G, Messer J, Waldvogel HJ, Gibbons HM, Dragunow M, Faull RL, et al. Up-regulation of the isoenzymes MAO-A and MAO-B in the human basal ganglia and pons in Huntington's disease revealed by quantitative enzyme radioautography. Brain Res. 2011 Jan 25;1370:204-14.

90. Chun H, Im H, Kang YJ, Kim Y, Shin JH, Won W, et al. Severe reactive astrocytes precipitate pathological hallmarks of Alzheimer's disease via H2O2- production. Nat Neurosci. 2020 Dec;23(12):1555-66.

91. Maggiorani D, Manzella N, Edmondson DE, Mattevi A, Parini A, Binda C, et al. Monoamine Oxidases, Oxidative Stress, and Altered Mitochondrial Dynamics in Cardiac Ageing. Oxid Med Cell Longev. 2017;2017:3017947.

92. Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, et al. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol. 2023 Oct;97(10):2499-574.

93. Ahmad IM, Aykin-Burns N, Sim JE, Walsh SA, Higashikubo R, Buettner GR, et al. Mitochondrial O2*- and H2O2 mediate glucose deprivation-induced stress in human cancer cells. J Biol Chem. 2005 Feb 11;280(6):4254-63.

94. Chen Y, Qin C, Huang J, Tang X, Liu C, Huang K, et al. The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif. 2020 Mar;53(3):e12781.

95. Rose J, Brian C, Pappa A, Panayiotidis MI, Franco R. Mitochondrial Metabolism in Astrocytes Regulates Brain Bioenergetics, Neurotransmission and Redox Balance. Front Neurosci. 2020 Nov 5;14:536682.

96. Soucek T, Cumming R, Dargusch R, Maher P, Schubert D. The regulation of glucose metabolism by HIF-1 mediates a neuroprotective response to amyloid beta peptide. Neuron. 2003 Jul 3;39(1):43-56.

97. Yun HJ, Li M, Guo D, Jeon SM, Park SH, Lim JS, et al. AMPK-HIF-1α signaling enhances glucose-derived de novo serine biosynthesis to promote glioblastoma growth. J Exp Clin Cancer Res. 2023 Dec 15;42(1):340.

98. Saura J, Luque JM, Cesura AM, Da Prada M, Chan-Palay V, Huber G, et al. Increased monoamine oxidase B activity in plaque-associated astrocytes of Alzheimer brains revealed by quantitative enzyme radioautography. Neuroscience. 1994 Sep;62(1):15-30.

99. Turner DA, Adamson DC. Neuronal-astrocyte metabolic interactions: understanding the transition into abnormal astrocytoma metabolism. J Neuropathol Exp Neurol. 2011 Mar;70(3):167-76.

100. Edmondson DE. Hydrogen peroxide produced by mitochondrial monoamine oxidase catalysis: biological implications. Curr Pharm Des. 2014;20(2):155-60.

101. Hong Y, Boiti A, Vallone D, Foulkes NS. Reactive Oxygen Species Signaling and Oxidative Stress: Transcriptional Regulation and Evolution. Antioxidants (Basel). 2024 Mar 1;13(3):312.

102. Stefanatos R, Sanz A. The role of mitochondrial ROS in the aging brain. FEBS Lett. 2018 Mar;592(5):743-58.

103. Beucher L, Gabillard-Lefort C, Baris OR, Mialet-Perez J. Monoamine oxidases: A missing link between mitochondria and inflammation in chronic diseases ? Redox Biol. 2024 Nov;77:103393.

104. Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel). 2023 Feb 18;12(2):517.

105. Franzoni F, Scarfò G, Guidotti S, Fusi J, Asomov M, Pruneti C. Oxidative Stress and Cognitive Decline: The Neuroprotective Role of Natural Antioxidants. Front Neurosci. 2021 Oct 13;15:729757.

106. Ostadkarampour M, Putnins EE. Monoamine Oxidase Inhibitors: A Review of Their Anti-Inflammatory Therapeutic Potential and Mechanisms of Action. Front Pharmacol. 2021 Apr 30;12:676239.

107. Sekar PK, Thomas SM, Veerabathiran R. An overview of the role of monoamine oxidase-B in Parkinson’s disease: implications for neurodegeneration and therapy. Explor Neuroprot Ther. 2024 Jul 14;4(4):308-18.

108. Kouli, A.; Torsney, K.M.; Kuan, W.L. Parkinson's Disease: Etiology, Neuropathology, and Pathogenesis. In Parkinson's Disease: Pathogenesis and Clinical Aspects, Stoker, T.B., Greenland, J.C., Eds.; Brisbane (AU), 2018.

109. Guatteo E, Berretta N, Monda V, Ledonne A, Mercuri NB. Pathophysiological Features of Nigral Dopaminergic Neurons in Animal Models of Parkinson's Disease. Int J Mol Sci. 2022 Apr 19;23(9):4508.

110. DeMaagd G, Philip A. Parkinson's Disease and Its Management: Part 1: Disease Entity, Risk Factors, Pathophysiology, Clinical Presentation, and Diagnosis. P T. 2015 Aug;40(8):504-32.

111. Maiti P, Manna J, Dunbar GL. Current understanding of the molecular mechanisms in Parkinson's disease: Targets for potential treatments. Transl Neurodegener. 2017 Oct 25;6:28.

112. Jankovic J, Tan EK. Parkinson's disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020 Aug;91(8):795-808.

113. Robakis D, Fahn S. Defining the Role of the Monoamine Oxidase-B Inhibitors for Parkinson's Disease. CNS Drugs. 2015 Jun;29(6):433-41.

114. Tan YY, Jenner P, Chen SD. Monoamine Oxidase-B Inhibitors for the Treatment of Parkinson's Disease: Past, Present, and Future. J Parkinsons Dis. 2022;12(2):477-93.

115. Muddapu VR, Chakravarthy VS. Influence of energy deficiency on the subcellular processes of Substantia Nigra Pars Compacta cell for understanding Parkinsonian neurodegeneration. Sci Rep. 2021 Jan 18;11(1):1754.

116. Teo KC, Ho SL. Monoamine oxidase-B (MAO-B) inhibitors: implications for disease-modification in Parkinson's disease. Transl Neurodegener. 2013 Sep 8;2(1):19.

117. Naoi M, Maruyama W, Shamoto-Nagai M. Neuroprotective Function of Rasagiline and Selegiline, Inhibitors of Type B Monoamine Oxidase, and Role of Monoamine Oxidases in Synucleinopathies. Int J Mol Sci. 2022 Sep 21;23(19):11059.

118. Alborghetti M, Bianchini E, De Carolis L, Galli S, Pontieri FE, Rinaldi D. Type-B monoamine oxidase inhibitors in neurological diseases: clinical applications based on preclinical findings. Neural Regen Res. 2024 Jan;19(1):16-21.

119. Karahoda R, Horackova H, Kastner P, Matthios A, Cerveny L, Kucera R, et al. Serotonin homeostasis in the materno-foetal interface at term: Role of transporters (SERT/SLC6A4 and OCT3/SLC22A3) and monoamine oxidase A (MAO-A) in uptake and degradation of serotonin by human and rat term placenta. Acta Physiol (Oxf). 2020 Aug;229(4):e13478.

120. Shih JC. Monoamine oxidase isoenzymes: genes, functions and targets for behavior and cancer therapy. J Neural Transm (Vienna). 2018 Nov;125(11):1553-66.

121. Jones DN, Raghanti MA. The role of monoamine oxidase enzymes in the pathophysiology of neurological disorders. J Chem Neuroanat. 2021 Jul;114:101957.

122. Yu YW, Tsai SJ, Hong CJ, Chen TJ, Chen MC, Yang CW. Association study of a monoamine oxidase a gene promoter polymorphism with major depressive disorder and antidepressant response. Neuropsychopharmacology. 2005 Sep;30(9):1719-23.

123. Thöny B, Ng J, Kurian MA, Mills P, Martinez A. Mouse models for inherited monoamine neurotransmitter disorders. J Inherit Metab Dis. 2024 May;47(3):533-50.

124. Kolla NJ, Bortolato M. The role of monoamine oxidase A in the neurobiology of aggressive, antisocial, and violent behavior: A tale of mice and men. Prog Neurobiol. 2020 Nov;194:101875.

125. Nam MH, Cho J, Kwon DH, Park JY, Woo J, Lee JM, et al. Excessive Astrocytic GABA Causes Cortical Hypometabolism and Impedes Functional Recovery after Subcortical Stroke. Cell Rep. 2020 Jul 7;32(1):107861.

126. An H, Heo JY, Lee CJ, Nam MH. The Pathological Role of Astrocytic MAOB in Parkinsonism Revealed by Genetic Ablation and Over-expression of MAOB. Exp Neurobiol. 2021 Apr 30;30(2):113-9.

127. Nam MH, Park JH, Song HJ, Choi JW, Kim S, Jang BK, et al. KDS2010, a Newly Developed Reversible MAO-B Inhibitor, as an Effective Therapeutic Candidate for Parkinson's Disease. Neurotherapeutics. 2021 Jul;18(3):1729-47.

128. Verkhratsky A, Butt A, Li B, Illes P, Zorec R, Semyanov A, et al. Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct Target Ther. 2023 Oct 13;8(1):396.

129. Heo JY, Nam MH, Yoon HH, Kim J, Hwang YJ, Won W, et al. Aberrant Tonic Inhibition of Dopaminergic Neuronal Activity Causes Motor Symptoms in Animal Models of Parkinson's Disease. Curr Biol. 2020 Jan 20;30(2):276-291.e9.

130. Tong J, Rathitharan G, Meyer JH, Furukawa Y, Ang LC, Boileau I, et al. Brain monoamine oxidase B and A in human parkinsonian dopamine deficiency disorders. Brain. 2017 Sep 1;140(9):2460-74.

131. Nagatsu T. Progress in monoamine oxidase (MAO) research in relation to genetic engineering. Neurotoxicology. 2004 Jan;25(1-2):11-20.

132. Arai R, Horiike K, Hasegawa Y. Dopamine-degrading activity of monoamine oxidase is not detected by histochemistry in neurons of the substantia nigra pars compacta of the rat. Brain Res. 1998 Nov 23;812(1-2):275-8.

133. Lamensdorf I, Youdim MB, Finberg JP. Effect of long-term treatment with selective monoamine oxidase A and B inhibitors on dopamine release from rat striatum in vivo. J Neurochem. 1996 Oct;67(4):1532-9.

134. Naoi M, Riederer P, Maruyama W. Modulation of monoamine oxidase (MAO) expression in neuropsychiatric disorders: genetic and environmental factors involved in type A MAO expression. J Neural Transm (Vienna). 2016 Feb;123(2):91-106.

135. Sanfilippo C, Castrogiovanni P, Imbesi R, Lazzarino G, Di Pietro V, Li Volti G, et al. Sex-dependent monoamine oxidase isoforms expression patterns during human brain ageing. Mech Ageing Dev. 2021 Jul;197:111516.

136. Lin YC, Chang YT, Campbell M, Lin TP, Pan CC, Lee HC, et al. MAOA-a novel decision maker of apoptosis and autophagy in hormone refractory neuroendocrine prostate cancer cells. Sci Rep. 2017 Apr 12;7:46338.

137. Choi JY, Yun J, Hwang CJ, Lee HP, Kim HD, Chun H, et al. (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) Phenol Ameliorates MPTP-Induced Dopaminergic Neurodegeneration by Inhibiting the STAT3 Pathway. Int J Mol Sci. 2019 May 29;20(11):2632.

138. Ramsay RR. Molecular aspects of monoamine oxidase B. Prog Neuropsychopharmacol Biol Psychiatry. 2016 Aug 1;69:81-9.

139. Chenu F, El Mansari M, Blier P. Long-term administration of monoamine oxidase inhibitors alters the firing rate and pattern of dopamine neurons in the ventral tegmental area. Int J Neuropsychopharmacol. 2009 May;12(4):475-85.

140. Cai Z. Monoamine oxidase inhibitors: promising therapeutic agents for Alzheimer's disease (Review). Mol Med Rep. 2014 May;9(5):1533-41.

141. Finberg JP, Rabey JM. Inhibitors of MAO-A and MAO-B in Psychiatry and Neurology. Front Pharmacol. 2016 Oct 18;7:340.

142. Zeng XS, Geng WS, Jia JJ. Neurotoxin-Induced Animal Models of Parkinson Disease: Pathogenic Mechanism and Assessment. ASN Neuro. 2018 Jan-Dec;10:1759091418777438.

143. Meredith GE, Rademacher DJ. MPTP mouse models of Parkinson's disease: an update. J Parkinsons Dis. 2011;1(1):19-33.

144. Onofrj M, Ghilardi MF. MPTP induced parkinsonian syndrome: long term follow-up and neurophysiological study. Ital J Neurol Sci. 1990 Oct;11(5):445-58.

145. AlShimemeri S, Di Luca DG, Fox SH. MPTP Parkinsonism and Implications for Understanding Parkinson's Disease. Mov Disord Clin Pract. 2021 Sep 25;9(1):42-7.

146. Kupsch A, Sautter J, Götz ME, Breithaupt W, Schwarz J, Youdim MB, et al. Monoamine oxidase-inhibition and MPTP-induced neurotoxicity in the non-human primate: comparison of rasagiline (TVP 1012) with selegiline. J Neural Transm (Vienna). 2001;108(8-9):985-1009.

147. Przedborski S, Tieu K, Perier C, Vila M. MPTP as a mitochondrial neurotoxic model of Parkinson's disease. J Bioenerg Biomembr. 2004 Aug;36(4):375-9.

148. Zhang YM, Qi YB, Gao YN, Chen WG, Zhou T, Zang Y, et al. Astrocyte metabolism and signaling pathways in the CNS. Front Neurosci. 2023 Sep 4;17:1217451.

149. Lee JM, Sa M, An H, Kim JMJ, Kwon J, Yoon BE, et al. Generation of Astrocyte-Specific MAOB Conditional Knockout Mouse with Minimal Tonic GABA Inhibition. Exp Neurobiol. 2022 Jun 30;31(3):158-72.

150. Edison P. Astroglial activation: Current concepts and future directions. Alzheimers Dement. 2024 Apr;20(4):3034-53.

151. Jia F, Pignataro L, Schofield CM, Yue M, Harrison NL, Goldstein PA. An extrasynaptic GABAA receptor mediates tonic inhibition in thalamic VB neurons. J Neurophysiol. 2005 Dec;94(6):4491-501.

152. Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev. 2018 Jan 1;98(1):239-389.

153. Chun H, Lim J, Park KD, Lee CJ. Inhibition of monoamine oxidase B prevents reactive astrogliosis and scar formation in stab wound injury model. Glia. 2022 Feb;70(2):354-67.

154. Rausch WD, Wang F, Radad K. From the tyrosine hydroxylase hypothesis of Parkinson's disease to modern strategies: a short historical overview. J Neural Transm (Vienna). 2022 Jun;129(5-6):487-95.

155. Henrich MT, Oertel WH, Surmeier DJ, Geibl FF. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential. Mol Neurodegener. 2023 Nov 11;18(1):83.

156. Dias V, Junn E, Mouradian MM. The role of oxidative stress in Parkinson's disease. J Parkinsons Dis. 2013;3(4):461-91.

157. Cao X, Wei Z, Gabriel GG, Li X, Mousseau DD. Calcium-sensitive regulation of monoamine oxidase-A contributes to the production of peroxyradicals in hippocampal cultures: implications for Alzheimer disease-related pathology. BMC Neurosci. 2007 Sep 16;8:73.

158. Finberg JP. Update on the pharmacology of selective inhibitors of MAO-A and MAO-B: focus on modulation of CNS monoamine neurotransmitter release. Pharmacol Ther. 2014 Aug;143(2):133-52.

159. Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, et al. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol. 2025 Feb;62(2):1631-74.

160. Park MG, Lim J, Kim D, Lee WS, Yoon BE, Lee CJ. Suppressing astrocytic GABA transaminase enhances tonic inhibition and weakens hippocampal spatial memory. Exp Mol Med. 2025 Feb;57(2):379-89.

161. Ju YH, Bhalla M, Hyeon SJ, Oh JE, Yoo S, Chae U, et al. Astrocytic urea cycle detoxifies Aβ-derived ammonia while impairing memory in Alzheimer's disease. Cell Metab. 2022 Aug 2;34(8):1104-1120.e8.

162. Emilsson L, Saetre P, Balciuniene J, Castensson A, Cairns N, Jazin EE. Increased monoamine oxidase messenger RNA expression levels in frontal cortex of Alzheimer's disease patients. Neurosci Lett. 2002 Jun 21;326(1):56-60.

163. Alborghetti M, Nicoletti F. Different Generations of Type-B Monoamine Oxidase Inhibitors in Parkinson's Disease: From Bench to Bedside. Curr Neuropharmacol. 2019;17(9):861-73.

164. Kim S, Chun H, Kim Y, Kim Y, Park U, Chu J, et al. Astrocytic autophagy plasticity modulates Aβ clearance and cognitive function in Alzheimer's disease. Mol Neurodegener. 2024 Jul 23;19(1):55.

165. Roth FC, Draguhn A. GABA metabolism and transport: effects on synaptic efficacy. Neural Plast. 2012;2012:805830.

166. Wen Y, Dong Z, Liu J, Axerio-Cilies P, Du Y, Li J, et al. Glutamate and GABAA receptor crosstalk mediates homeostatic regulation of neuronal excitation in the mammalian brain. Signal Transduct Target Ther. 2022 Oct 3;7(1):340.

167. Beckhauser TF, Francis-Oliveira J, De Pasquale R. Reactive Oxygen Species: Physiological and Physiopathological Effects on Synaptic Plasticity. J Exp Neurosci. 2016 Sep 4;10(Suppl 1):23-48.

168. Massaad CA, Klann E. Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid Redox Signal. 2011 May 15;14(10):2013-54.

169. Beltrán González AN, López Pazos MI, Calvo DJ. Reactive Oxygen Species in the Regulation of the GABA Mediated Inhibitory Neurotransmission. Neuroscience. 2020 Jul 15;439:137-45.

170. Xu Y, Zhao M, Han Y, Zhang H. GABAergic Inhibitory Interneuron Deficits in Alzheimer's Disease: Implications for Treatment. Front Neurosci. 2020 Jun 30;14:660.

171. Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol. 2021 Aug;116:146-59.

172. Sun Z, Sun L, Tu L. GABAB Receptor-Mediated PI3K/Akt Signaling Pathway Alleviates Oxidative Stress and Neuronal Cell Injury in a Rat Model of Alzheimer's Disease. J Alzheimers Dis. 2020;76(4):1513-26.

173. Li Y, Sun H, Chen Z, Xu H, Bu G, Zheng H. Implications of GABAergic Neurotransmission in Alzheimer's Disease. Front Aging Neurosci. 2016 Feb 23;8:31.

174. 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 Dec;92:102123.

175. Tamagno E, Guglielmotto M, Vasciaveo V, Tabaton M. Oxidative Stress and Beta Amyloid in Alzheimer's Disease. Which Comes First: The Chicken or the Egg? Antioxidants (Basel). 2021 Sep 16;10(9):1479.

176. Roy RG, Mandal PK, Maroon JC. Oxidative Stress Occurs Prior to Amyloid Aβ Plaque Formation and Tau Phosphorylation in Alzheimer's Disease: Role of Glutathione and Metal Ions. ACS Chem Neurosci. 2023 Sep 6;14(17):2944-54.

177. Diep YN, Park HJ, Kwon JH, Tran M, Ko HY, Jo H, et al. Astrocytic scar restricting glioblastoma via glutamate-MAO-B activity in glioblastoma-microglia assembloid. Biomater Res. 2023 Jul 19;27(1):71.

178. Yang T, Dai Y, Chen G, Cui S. Dissecting the Dual Role of the Glial Scar and Scar-Forming Astrocytes in Spinal Cord Injury. Front Cell Neurosci. 2020 Apr 3;14:78.

179. Yu G, Zhang Y, Ning B. Reactive Astrocytes in Central Nervous System Injury: Subgroup and Potential Therapy. Front Cell Neurosci. 2021 Dec 23;15:792764.

180. Haskew-Layton RE, Payappilly JB, Smirnova NA, Ma TC, Chan KK, Murphy TH, et al. Controlled enzymatic production of astrocytic hydrogen peroxide protects neurons from oxidative stress via an Nrf2-independent pathway. Proc Natl Acad Sci U S A. 2010 Oct 5;107(40):17385-90.

181. Lawrence JM, Schardien K, Wigdahl B, Nonnemacher MR. Roles of neuropathology-associated reactive astrocytes: a systematic review. Acta Neuropathol Commun. 2023 Mar 13;11(1):42.

182. Alsaad I, Abdel Rahman DMA, Al-Tamimi O, Alhaj SA, Sabbah DA, Hajjo R, et al. Targeting MAO-B with Small-Molecule Inhibitors: A Decade of Advances in Anticancer Research (2012-2024). Molecules. 2024 Dec 31;30(1):126.

183. Hâncu IM, Giuchici S, Furdui-Lința AV, Lolescu B, Sturza A, Muntean DM, et al. The highs and lows of monoamine oxidase as molecular target in cancer: an updated review. Mol Cell Biochem. 2024 Dec 23.

184. Zhao Y, Ye X, Xiong Z, Ihsan A, Ares I, Martínez M, et al. Cancer Metabolism: The Role of ROS in DNA Damage and Induction of Apoptosis in Cancer Cells. Metabolites. 2023 Jun 27;13(7):796.

185. Resta J, Santin Y, Roumiguié M, Riant E, Lucas A, Couderc B, et al. Monoamine Oxidase Inhibitors Prevent Glucose-Dependent Energy Production, Proliferation and Migration of Bladder Carcinoma Cells. Int J Mol Sci. 2022 Oct 4;23(19):11747.

186. Sharpe MA, Raghavan S, Baskin DS. PAM-OBG: A monoamine oxidase B specific prodrug that inhibits MGMT and generates DNA interstrand crosslinks, potentiating temozolomide and chemoradiation therapy in intracranial glioblastoma. Oncotarget. 2018 May 8;9(35):23923-43.

187. Pu T, Wang J, Wei J, Zeng A, Zhang J, Chen J, et al. Stromal-derived MAOB promotes prostate cancer growth and progression. Sci Adv. 2024 Feb 9;10(6):eadi4935.

188. Andersen JV. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J Neurochem. 2025 Mar;169(3):e70029.

189. Xie M, Qin H, Liu L, Wu J, Zhao Z, Zhao Y, et al. GABA regulates metabolic reprogramming to mediate the development of brain metastasis in non-small cell lung cancer. J Exp Clin Cancer Res. 2025 Feb 19;44(1):61.

190. Yang Y, Ren L, Li W, Zhang Y, Zhang S, Ge B, et al. GABAergic signaling as a potential therapeutic target in cancers. Biomed Pharmacother. 2023 May;161:114410.

191. Huang D, Alexander PB, Li QJ, Wang XF. GABAergic signaling beyond synapses: an emerging target for cancer therapy. Trends Cell Biol. 2023 May;33(5):403-12.

Author Information X