Abstract
Memory is a dynamic process of encoding, storing, and retrieving information. It includes sensory, short-term, and long-term memory, each with unique characteristics. Nitric oxide (NO) is a biological messenger synthesized on demand by neuronal nitric oxide synthase (nNOS) through a biochemical process initiated by glutamate binding to NMDA receptors, causing membrane depolarization and calcium influx. NO is known to regulate many signaling pathways including those related to memory consolidation. To throw light on the precise molecular mechanism of nitric oxide (NO) in memory consolidation and the possibility of targeting NO pathways as a therapeutic approach to scale down cognitive impairments. We conducted a search of the PubMed MEDLINE database, maintained by the US National Library of Medicine. The search strategy utilized Medical Subject Headings (MeSH) terms, including “nitric oxide and memory,” “nitric oxide synthesis in the brain,” “nitric oxide and Alzheimer’s,” “nitric oxide and Parkinson’s,” and “nitric oxide, neurodegenerative disorders, and psychiatric disorders.” Additionally, relevant keywords such as “nitric oxide,” “memory,” and “cognitive disorders” were employed. We included the most recent preclinical and clinical studies pertinent to the review topic and limited the selection to articles published in English. NO exerts its role in memory consolidation by diffusing between neurons to promote synaptic plasticity, especially long-term potentiation (LTP). It acts as a retrograde messenger, neurotransmitter release modulator, and synaptic protein modifier. The dysregulation of NO balance in the brain can contribute to the pathogenesis of various neurodegenerative diseases, particularly Alzheimer’s, Parkinson’s, and psychiatric disorders. The disturbance in NO signaling is strongly correlated with synaptic signaling dysfunction and oxidative stress. NO plays a fundamental role in memory consolidation, and its dysregulation contributes to cognitive impairment—a hallmark of numerous neurodegenerative and psychiatric disorders. Future research should aim to deepen our understanding of the mechanisms underlying NO’s involvement in memory consolidation and to explore therapeutic strategies targeting the NO pathway to mitigate cognitive decline in affected individuals.
Keywords: Nitric oxide, Short-term memory, Long-term memory, Molecular mechanism, Pathogenesis
Introduction
Memory is a dynamic chemical process between neurons [1, 2]. Many scientists have concurred to define memory as the cognitive ability that allows the human high brain centers to encode, store, and retrieve information when needed; it is the perpetuation of information for future influencing act [3], like adapting, learning, solving problems, and making decisions. According to the Atkinson–Shiffrin memory model (Fig. 1) that was proposed in 1968, information that enters our brain and forms a memory passes through three co-depended stages; sensory memory, short-term memory (STM), and long-term memory (LTM) [4].
Fig. 1.
The Atkinson–Shiffrin memory model. This model describes the three stages of memory formation: sensory memory, short-term memory, and long-term memory. It also highlights the importance of rehearsal to pass through these stages and form a long-term, life-long memory
Sensory memory refers to the memory formed for sensorial details of a life situation, this includes any detail that can be obtained by our five senses: visual, auditorial, tactile, olfactory, and gustatorial [5]. It allows us to recognize a familiar scene, voice, or the face of someone we know. It can accurately receive this information but ultimately fades away [6]. Although this memory has a great capacity, these sensorial details last only for milliseconds [6] and are mostly outside our conscious awareness as these memories are captured through the auditorium and visual pathways, and then sent to be relayed on the thalamus through the sensory cortices [7, 8].
Short-term memory (STM) refers to the retention of details and information in conscious awareness for only a few seconds, and it is limited in capacity. In fact, an erudite theory developed by Miller in the 1950s states that short-term memory holds seven units of information, plus or minus two items, although new research suggested that it is closer to four items, and other research proposed that it is reliant on the individual’s processing speed [9]. The information obtained in short-term memory will eventually leave the processing system leading to either an evanescent response or being stored in the long-term memory [9]. The Atkinson and Shiffrin model suggested that unless we rehearse the obtained information verbally, the information can stay in the STM for 15–30 s before we start losing it [6, 10].
LTM refers to the memory that is held for a longer period or considered everlasting It has an enormous capacity that store complicated knowledge structured into cognitive dynamic schemas, which can be alternatively defined as distinct pieces of obtained knowledge that organize and categorize the data [6]. The LTM is primarily the function of the hippocampus and is stored throughout the cortex [11]. Long-term memories can be classified into two categories: procedural (or implicit) and declarative (or explicit). Procedural memory is the memory that forms by implementing and repeating exposure to a series of motor outputs [11]. Declarative memory is the process of forming intentional memories by the direct recollection of event content and can be further sub-classified as episodic memory about personal experiences and their related details and semantic memory about factual knowledge [12].
NO is a small and highly diffusible neurotransmitter that acts as a biological messenger. It possesses many unique characteristics and participates in many cellular signaling pathways. It is not synthesized after membrane depolarization and is not stored in synaptic vesicles like many neurotransmitters, nevertheless, it is made and released upon cellular needs. In addition to that, NO doesn’t perform its function by binding to receptors on the targeted membrane, but by diffusing between the neurons and acting directly on the cell’s intracellular components [13]. This potent molecule plays an essential role in many physiological processes within the body cells, particularly in the nervous system as a neurotransmitter and neuromodulator. In the central nervous system (CNS), nitric oxide has different roles, including the regulation of synaptic plasticity, the sleep-wake cycle, and hormone secretion [14]. It largely plays a vital role in enhancing the functionality of the hypothalamus pertaining to the learning process and memory formation [13, 15].
To obtain the desired level of NO, a series of biochemical steps must be meticulously followed. NO synthesis starts with Glutamate binding to the NMDA receptor which will eventually cause a membrane depolarization and the influx of calcium. Calcium will bind to calmodulin forming the co-factor complex; the calcium-calmodulin cofactor, which activates the nitric oxide synthase (NOS). NOS then acts on the primary substrate L-arginine, which is taken up by the cationic amino acid transporter (CAT-1). L-arginine will then be hydroxylated first into N-hydroxy-L-arginine, and then oxidized into two molecules: L-citrulline and nitric oxide [13].
The recognition of nitric oxide’s role in memory consolidation is vital, thus it is considered the mainspring in understanding the physiology of cognition and an important target for current and future therapeutic applications. Despite the continuous advancement in comprehending many aspects in the field of neuroscience, there are still gaps that need to be filled about the precise mechanism by which NO influences memory consolidation, or what are the conditions by which NO exerts its effect. Based on that our minireview aims to emphasize the intriguing implication of nitric oxide in memory formation and the impact of NO dysregulation in memory-related diseases. This review looks further at how to use nitric oxide as a target for treating or reducing the disease severity in memory-related disorders such as Alzheimer’s disease, and other forms of cognitive impairment related to psychiatric disorders.
Nitric oxide formation and its mechanism in memory consolidation
Nitric oxide formation
Nitric oxide has multiple regulatory functions in our central nervous system, ranging from vasodilation to neurotransmission, and immune defense mechanisms. Nitric oxide synthesis in our central nervous system is primarily catalyzed by the nitric oxide synthase (NOS). The NOS has three isoforms: endothelial NOS (eNOS), neuron NOS (nNOS), and inflammatory NOS (iNOS). Despite that both eNOS and iNOS are present in the CNS, nNOS is the major enzyme responsible for neuronal NO production [16]. nNOS gene is regulated at the transcriptional level. Sp1 and CREB both regulate the initiation of gene transcription in response to the activation of neurons. This transcription will lead to the translation of mRNA into the nNOS in the cytoplasm, where it will undergo posttranslational modification. nNOS will interact with the calcium calmodulin complex, which was previously formed when glutamate is bound to the NMDA receptor, allowing depolarization of the membrane, and causing an influx of calcium. To get nitric oxide as a by-product, we start with the substrate L-arginine that enters the cytoplasm through the cationic amino acid transporter (CAT1) transporter. nNOS will interact with L-arginine causing hydroxylation into N-hydroxy-L-arginine and then oxidized with the aid of the co-factor tetrahydrobiopterin (BH4) that couples to the nNOS enzyme to get two products: L-citrulline and nitric oxide. Thus, the absence of the substrate or BH4 does not result in the production of nitric oxide [17]. This absence might be caused by conditions that increase oxidative stress, such as UV radiation and ionizing radiation, resulting in NOS uncoupling and the generation of highly oxidative free radicals [18]. Other co-factors include NADPH, heme, Flavin Adenine Dinucleotide (FAD), and Flavin Mononucleotide (FMN) all involved in transferring electrons within the enzyme complex, thus facilitating the conversion of L-arginine to nitric oxide (NO) and L-citrulline. The nitric oxide will then diffuse to the postsynaptic cleft to exert its multiple effects on nearby neurons [19]. The biochemical steps of Nitric oxide synthesis and the factors required to speed up nitric oxide production have been illustrated in Fig. 2.
Fig. 2.
The biochemical steps for NO synthesis and the important molecular factors required for its production
Nitric oxide mechanism in memory consolidation
Memory consolidation is defined as the complex process in which the temporary volatile memory is changed into a more stable, long-lasting form [20]. Nitric oxide plays a pivotal role in memory consolidation, via its involvement in synaptic plasticity, a fundamental process in which the connection between the neurons can strengthen or weaken over time according to their activity, which is crucial for encoding and tracing storage of the type of memory mediated by the brain area in which is observed [21]. Nitric oxide participates in synaptic plasticity through enhancing long-term potentiation (LTP) in the hippocampus and cerebral cortex [22].
Nitric oxide affects synaptic plasticity through different pathways to facilitate this transmission of memories into a more stable, permeant form. it functions as a retrograde messenger, modulating neurotransmitter release, regulating the dynamics of synaptic proteins, and impacting gene expression and protein synthesis, NO ensures that neural circuits are optimally configured for the long-term retention of information. This multi-pathway clears the air about how nitric oxide is pivotal in cognitive functions and its potential for future therapeutic targeting strategies in treating memory-related disorders [23].
Nitric oxide as a retrograde messenger
Nitric oxide serves as a retrograde messenger after postsynaptic NMDA (N-methyl-D-aspartate) receptor activation in hippocampal long-term potentiation (LTP) [22]. It all starts when the NMDA receptors on the postsynaptic are activated, leading to the influx of calcium ions (Ca + 2) [24]. This influx activates neuronal NOS leading to synthesizing the final product nitric NO. NO then diffuses rapidly to the presynaptic cleft where soluble guanylate cyclase (sGC) is activated. sGC catalyzes the conversion of GTP to cyclic GMP (cGMP), where the increase in cGMP causes the activation of protein kinase G [25]. Protein kinase G (PKG) in turn phosphorylates multiple proteins involved in NO release, such as glutamate, thus enhancing the synaptic strength and promoting LTP. The abovementioned complicated signaling cascade emphasizes the importance of NO in regulating synaptic plasticity and memory consolidation [25].
Nitric oxide modulation of S-nitrosylation of proteins
Nitric oxide also plays a role in modulating neurotransmitter release through various pathways, one of which involves modulating the release through the S-nitrosylation of proteins. S-nitrosylation is a post-translational modification of proteins in which the nitric oxide is covalently bound to the free sulfhydryl group of specific cysteine residues of the targeted protein, resulting in the formation of S–S-nitrosothiol (SNOI) [26]. For instance, S-nitrosylation can impact protein-protein interactions, either promoting or inhibiting the binding of proteins to their cognate partners. The S-nitrosylation of NMDA (N-methyl-D-aspartate) receptors were found to affect the calcium influx, therefore having an impact on the learning and memory process [27]. The enhancement of the NMDA receptor’s activity via S-nitrosylation will cause an increase in calcium influx, promoting synaptic plasticity [28]. The S-nitrosylation has a dual role of being neuroprotective and neurotoxic. It can protect the nervous system by inhibiting harmful processes such as excessive NMDA receptor activity, which can lead to excitotoxicity and neuronal damage [2]. Meanwhile, an abnormal concentration of S-nitrosylation can interfere with mitochondrial function leading to impaired energy production, synaptic damage, and increased oxidative stress, and collectively can cause neurotoxic effects which are implicated in multiple neurological disorders [2, 29]. Nitric oxide-mediated S-nitrosylation enhances NMDA receptor stimulation, thereby promoting calcium influx and synaptic plasticity, both of which are required for memory retention and retrieval [30]. A study by Forrester et al. (2009) described the biotin-switch technique (BST), a commonly used assay for detecting S-nitrosylated proteins in complicated biological systems. This approach has proved useful in understanding the conducted role of nitric oxide in modifying protein S-nitrosylation, which influences a variety of cellular processes such as neurotransmitter release and synaptic plasticity. BST has shed light on the molecular mechanisms governing memory preservation and retrieval by allowing the identification and quantification of S-nitrosylated proteins [31]. Building upon the role of NO in modulating synaptic activity through S-nitrosylation, it is also known to influence the activity of calcium/calmodulin-dependent protein kinase II (CaMKII), a major enzyme involved in synaptic plasticity and memory processes. One study found that NO suppresses CaMKII activity via S-nitrosylation, affecting synaptic function and memory functions [32]. This study found that reducing agents can reverse NO-mediated inhibition of CaMKII, showing that S-nitrosylation is reversible and NO plays a dynamic function in controlling synaptic plasticity.
Nitric oxide modulation of Calcium/Calmodulin-Dependent kinases (CaMKs)
CaMK (calcium calmodulin-dependent kinase) is a major holoenzyme of the postsynaptic density that applies its function through phosphorylating key receptors such as the NMDA and AMPAR [33]. Upon calcium influx, CaMK undergoes autophosphorylation, resulting in prolonged Ca2+/CaM-independent autonomous kinase activity, which brings out the prolonged enhancement of LTP [34]. Notably, NO-cGMP signaling pathways intersect with CaMK pathways, further influencing their activity and contributing to the highest, intricate levels of neuronal signaling and plasticity mechanisms [35]. Furthermore, this calcium influx activates calcium/calmodulin-dependent protein kinase II (CaMKII), a multifunctional enzyme abundant in glutamatergic synapses, significantly contributing to calcium signal transduction, synaptic function, and memory-related processes [36]. This modulation is critical for sustaining the synaptic alterations that are crucial for underpinning memory retention.
Nitric oxide regulation of synaptic proteins
Nitric oxide can influence the activity of protein kinases and phosphatases to regulate the phosphorylation status of important proteins and transcriptional factors, importantly CREB (cAMP response element-binding protein), which regulates genes associated with synaptic development and plasticity [37]. To identify the role of CREB in memory consolidation, scientists studied mutated mice with genetic loss of CREB function, in which the formation of LTM was affected without disturbing STM [1]. CREB functions by targeting the transcription of neuronally important genes, including c-fos, activity-regulated cytoskeleton-associated protein (Arc), and brain-derived neurotrophic factors (BDNF). It has been suggested that CREB regulates STM via the activation of above-listed genes, such as BDNF [38, 39]. The Morris water maze, a hippocampal-dependent spatial memory task, has shown that nitric oxide (NO) signaling deficiencies impair long-term potentiation (LTP) and reduce CREB activation via protein kinase A alterations, resulting in profound deficits in the consolidation of long-term spatial memories, emphasizing the critical interplay between NO, CREB, and hippocampal synaptic plasticity in learning and memory [40].
Nitric oxide impact on AMPA receptors
AMPA receptors (AMPARs) are glutamate-gated ion channels, and their main function is to mediate fast excitatory synaptic transmission [41]. Nitric oxide facilitates the insertion of AMPARs into the postsynaptic membrane, indirectly by activating the cGMP-dependent cascade [42]. The NO-cGMP-PKG (cyclic guanosine monophosphate-protein kinase G) signaling pathway possibly promotes synaptic plasticity and fear memory formation by activating the ERK/MAPK signaling cascade [43]. NO activates soluble guanylate cyclase (sGC), increasing cGMP levels, and activating PKG. The activation of PKG facilitates the incorporation of AMPAR postsynaptically, thus increasing the number of receptors available for synaptic transmission to expedite the LTP [42, 43]. Pavlovian fear conditioning is conducted via This test involves pairing a neutral stimulus with an aversive stimulus to establish an associative memory, where this test shows that Reduced NO signaling has been shown to impair memory retrieval and synaptic plasticity, including AMPA receptor-mediated activity, in such paradigms [44].
Crosstalk between NO and other gasotransmitters in memory processes
CO (carbon monoxide), H2S (hydrogen disulfide), and NO (nitric oxide) are endogenous signaling molecules called gasotransmitters that transmit chemical signals, which induce various physiological and biochemical changes. CO signaling is a molecular regulator of the NO feedback ring that can in turn stimulate CO synthesis by modulating the level of HO-1 protein, which in turn, can influence the formation of NO [45]. Both NO and CO play a role in synaptic plasticity and memory formation by activating the soluble guanylate cyclase (sGC) [46]. H2S also overlaps with NO signaling, complementing NO bioavailability by stabilizing nitrosothiol or reacting with reactive oxygen species (ROS) [46], thereby exerting its protective role as anti-apoptosis [45], which is crucial for regulating memory-related signaling pathways. Playing together, these Gasotransmitters engage in performing crucial role in memory and other neurophysiological processes by regulating vascular tone, inducing neuroprotection, and consolidating synaptic plasticity [46].
Experimental evidence supporting NO’s role in memory consolidation
Multiple studies have been conducted to provide supporting evidence that nitric oxide does have a role in memory consolidation. A pharmacological inhibitor of NO was used to evaluate whether NO-mediated LTP has an essential role in memory consolidation or not. The NOS inhibitors that have been assessed are N-nitro-L-arginine and NG-methyl-L-arginine and were used intracellularly and extracellularly. The obtained results revealed that NO is a crucial contributor to enhancing the hippocampal LTP by reinforcing the involved synapses and facilitating the memory circuits [47]. Another study evaluated the involvement of PKG in LTP by using specific inhibitors and activators to target this kinase. This study showed that PKG inhibitor blocked the LTP induction while the use of PKG activator produced activity-dependent long-lasting enhancement suggesting that guanylyl cyclase and PKG are potent contributors to support the LTP. The activation of these molecules is tightly dependent on NO availability [48]. Another study used the Morris-water maze (MWM) in mice lacking nNOS (knocked-out) to study the learning and memory functions and cognitive abilities. Most of the nNOS knocked-out mice failed to find the submerged platform within the given time indicating that these mice had impaired spatial performance due to the direct effects of nNOS dysfunction [49].
NO-Mediated epigenetic modifications in memory consolidation
NO plays a vital role in regulating epigenetic modifications, which are vital for memory consolidation. One mechanism is facilitating the dissociation of histone deacetylase 2 (HDAC2) from CREB-regulated gene promoters, via the activation of histone acetylation [50]. HDAC inhibitors can be used to increase histone H3 acetylation, by setting back the deficit observed in nitric oxide synthase (NOS) knockout mice [51]. Histone acetylation can overlap with the DNA methylation pathway. This is observed in impaired DNA methylation in the hippocampus due to NMDA receptor antagonists proposing a possible involvement of NO-relating signaling pathways in coordinating these epigenetic processes [52]. Additionally, the NO can affect chromatin remodeling in neurons and S-nitrosylation of HDAC2, which are essential for activity-dependent gene regulation during neural development and learning [52].
Nitric oxide dysregulation and memory-related disorders
Nitric oxide has a dual role of being neuroprotective at physiological levels and neurotoxic when being overproduced [53]. Thus, there is no doubt that any dysregulation in nitric oxide levels would result in severe neurodegenerative and psychiatric disorders. Alzheimer’s disease (AD) is a distinct example of a neurodegenerative disorder where changes in NO signaling contribute to synaptic dysfunction and cognitive impairment [54]. Parkinson’s disease (PD) is another neurodegenerative condition that also exhibits malfunctioning NO signaling which causes defective motor and cognitive functions [55]. Additionally, psychiatric conditions, including schizophrenia [56] and depression [57] have been associated with abnormalities in NO pathways, emphasizing the molecule’s importance in brain function and memory.
In recent years, drugs targeting NO pathways have gained popularity. For example, selective iNOS inhibitors like 1400 W at 15 mg/kg per day for 2 weeks are being studied in preclinical trials for their potential to reduce neuroinflammation in Alzheimer’s disease [58]. Similarly, NO donors such as glyceryl trinitrate and cGMP pathway modulators such as sildenafil have shown a potential for improving memory and cognitive performance without altering amyloid burden in animal models and preliminary clinical trials [59]. Antioxidants such as N-acetylcysteine have been also investigated for their capacity to prevent nitrosative stress and the related cognitive impairment manifested in psychiatric disorders such as bipolar and major depression [60].
Alzheimer’s disease (AD)
Alzheimer’s disease (AD) dementia is defined as a particular onset and progression of age-related cognitive and functional decline with a specific neuropathology [61]. The dysregulation of nitric oxide associated with advanced age and vascular dysfunction are contributing risk factors for AD [62]. To have proper cognitive function, nitric oxide is needed for neuronal communication and synaptic plasticity. In AD, the dysregulation of nitric oxide leads to multiple pathological features such as exacerbation of amyloid beta (Aβ) plaques deposition, disruption of synaptic signaling and plasticity, and chronic Inflammation. The overproduction of Aβ plaques exacerbates when nitric oxide is dysregulated causing the upregulation in the level of reactive oxygen species (ROS), particularly peroxynitrite [63, 64]. The overproduction of ROS accelerates cellular damage and leads to synaptic dysfunction [65]. Microglia produce neurotoxic NO [66] when they are activated during an inflammatory response to extracellular Aβ plaque deposition in the brain. Increased quantities of Aβ plaques can directly attach to mitochondrial outer and inner membranes changing their dynamics and functions and causing an aberrant energy metabolism which ensued a synaptic function loss [65]. A chronic inflammation is accompanied with a sustained high level of induced NO (iNOS) resulting and abnormal level of NO which further contribute to the deposition of Aβ plaques [67]. Overproduction of NO can also aggravate the synaptic plasticity disruption by altering the NMDA receptors expression, in which excessive NMDA receptors activation would cause neurotoxicity and promote cell death [68]. This can cause synaptic loss and contribute to the memory deficit observed in AD. Collectively, these mechanisms highlight the importance of restoring nitric oxide homeostasis as a possible prevention strategy or curing approach for Alzheimer’s.
Parkinson’s disease
Defined as a chronic neurodegenerative disease mainly of the central nervous system that affects both motor and non-motor symptoms, such as tremors, rigidity, bradykinesia, cognitive deficits, and memory impairment. Initial studies have proposed that the dysregulation of NO signaling may play a role in the cognitive deficit in PD. Since NO plays a pivotal role in synaptic plasticity and the maintenance of LTP, these studies suggested that the dysregulation of NOS enzymes in PD patients might result in nitric oxide dysregulation which triggered a noticeable deficit in both the spatial and declarative memories that commodification observed in PD patients [69]. Correspondingly, postpartum studies performed in Parkinson’s patients’ brains and 1- methyl-4-phenyl-1,2,3,6- tetrahydropyridine MPTP-treated mice revealed that nitric oxide plays a significant part in PD, where MPTP-induced nigral dopaminergic neuronal death in Parkinson’s patients. Glial cells convert MPTP to MPP + to be utilized by dopaminergic neurons through dopamine transporters. Accumulation of MPP + hinders complex I of the mitochondrial electron transport chain causing DNA damage and activating PARP-1, ultimately leading to cell death. Numerous studies indicated that NO has a role in MPP+-induced DNA damage, PARP-1 activation, and cell death [70]. Similarly to AD, the formation of peroxynitrite can contribute to neuronal death and cognitive decline in the dopaminergic system and other brain regions involved in memory processing such as the hippocampus and cortex, in addition to inflammation that would activate the microglial, leading to upregulation of iNOS [71], hence exacerbating the memory deficit. Thus, therapeutic targeting of NO offers a possible strategy to alleviate cognitive decline in PD. Inhibiting nNOS and iNOS and applying antioxidants were demonstrated to relieve the effects of nitrosative stress in PD [72], suggesting that restoring NO homeostasis could improve cognitive outcomes.
Schizophrenia
Schizophrenia is characterized by having psychotic symptoms, social and occupational decline with concomitant cognitive impairment [73]. Studies indicated that individuals with schizophrenia exhibited a significant disturbance in NO levels in the brain structures (cerebellum, hypothalamus, hippocampus, striatum) and fluids [56]. Several mechanisms of NO dysregulation possibly contribute to memory deficit in schizophrenic patients including impaired synaptic plasticity, altered glutamate signaling, and oxidative stress. Impaired synaptic plasticity in schizophrenic patients might be attributed to mitochondrial dysfunction leading to multiple manifestations, including altered ROS level and nitric oxide production [74]. This dysregulation in NO can disturb the LTP, causing memory formation impairment, as well as the excess ROS would damage brain regions like the prefrontal cortex and hippocampus, which are vital for memory. NMDA dysfunction seems to be particularly relevant in schizophrenic patients which have been validated by using NMDA antagonists that produced symptoms resembling schizophrenia manifestations [75]. Dopamine is believed to play a central role in schizophrenia as its increase contributes to the positive symptoms of schizophrenia, while its decrease contributes to the negative symptoms. Dopamine was also linked to the process of memory encoding and consolidation, particularly by stimulating the dopamine receptors. Stimulation of dopamine receptors as part of a hippocampal–striatal–prefrontal loop orchestrates the formation of new memories [76]. Thus, dysregulation of dopamine due to nitric oxide dysfunction would result in producing various symptoms related to schizophrenia. In schizophrenia, the dysregulation of NO triggers a series of molecular and cellular cascades that contribute to memory impairments. Maintaining balanced NO signaling is crucial for reviving the cognitive function in schizophrenic patients and can be purported as a potential therapeutic target for schizophrenia..
Depression
Nitric oxide is known for its influence on mental health in several pathways. Nitric oxide has a vital role in the pathogenesis of major depressive disorder by modulating the level of norepinephrine, serotonin, dopamine, and glutamate. These major neurotransmitters are involved in the neurobiology of major depression [77] and their disturbance is linked mechanistically with memory dysfunction in depression. A clinical study confirmed that the production of NO is increased in depressed patients [78]. NO influences memory deficit in depression as it plays a role in impairing the process of neurogenesis by instigating a disturbance in the level of NMDA receptors and neurotransmitters and enkindling baneful levels of oxidative stress and neuroinflammation. Accumulated reports have indicated that depression may be linked to the lack of hippocampal neurogenesis, which is negatively influenced by neuronal nNOS-derived NO, thus suggesting that the overexpression of nNOS in the hippocampus is necessary for chronic stress-induced depression, and inhibiting nNOS signaling in the brain may mitigate these debilitating effects [79]. Moreover, the dysregulation of NO signaling can hinder the regenerative abilities of neural stem cells (NSCs) causing an exacerbation in cognitive impairment associated with depression. As mentioned before, NO modulation of NMDA receptors also contributes to the memory deficit observed in depression. A study exposed that the antidepressant-like effect of the yueju pill on mice through the inhibition of the NMDA/NO/cGMP pathway providesREF12 evidence that supported the importance of NMDA receptors in memory deficit found in depressed patients [80]. NO modulates the neurotransmitter levels of both serotonin and dopamine which are implicated in mood regulation. Tetrahydrobiopterin (BH4) is a vital enzymatic cofactor required for the synthesis of serotonin, dopamine, and NO; thus, its dysregulation has been documented in many pathological situations, including Alzheimer’s disease, Parkinson’s disease, and depression as it increased the oxidative stress and neuroinflammation which can worsen the memory deficit exhibited in depressed patients [81]. The dysregulation of NO through its interaction with the serotonin transporter (SERT) negatively affects serotonin uptake by reducing SERT’s cell surface localization, thereby causing further deterioration in the depressed symptoms due to low availability of serotonin [82].
Bipolar disorder
Nitric oxide is crucial for memory consolidation as it promotes LTP and retrograde communication in the hippocampus and prefrontal cortex [83]. Neuronal nitric oxide synthase (nNOS) regulates NO production, which modulates synaptic strength and creates lasting memory circuits [84]. In bipolar disorder, the NO signaling pathway is profoundly affected. A case-control study showed that bipolar disorder patients with mania had lower nitric oxide (NO) levels than healthy controls, but the difference was not statistically significant. The study found a modest negative association between NO levels and disease duration, which could have consequences for the neurobiology of bipolar disorder during manic episodes [85]. Abnormal NO production, either excessive or insufficient, can cause nitrosative stress and impede neuroplasticity [86]. Investigating the molecular imbalances in NO homeostasis may act as a link between neuroinflammatory processes and cognitive dysfunction in bipolar disorder, potentially leading to targeted therapeutic interventions..
PTSD
Nitric oxide (NO), predominantly through neuronal nitric oxide synthase (nNOS), influences synaptic plasticity by altering neurotransmitter release and LTD, as both are necessary for memory and learning processes. In PTSD, abnormal NO signaling, including excessive NO production and NO-induced post-translational modifications such as S-nitrosylation, impair these processes, contributing to defective neuroplasticity and the persistence of maladaptive fear responses [87]. Elevated stress levels related to PTSD cause dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, leading to altered glucocorticoid secretion, which can modulate stress-related physiological responses [88]. Neuronal nitric oxide synthase (nNOS) activation by calcium influx via NMDAR, is critical for the fear learning stages related to PTSD. Pharmacological interventions targeting nNOS have been demonstrated to limit fear acquisition and fear memory while improving extinction consolidation, making these drugs a prospective treatment options for PTSD patients [89].
Conclusion
NO plays a pivotal role in memory consolidation through different molecular pathways such as acting as a retrograde messenger, modulating neurotransmitter release, regulating synaptic proteins, and influencing gene expression and protein synthesis. Thus, its dysregulation can negatively impact the synaptic plasticity in multiple neurological diseases, such as Alzheimer’s disease, Parkinson’s disease, schizophrenia, depression, bipolar, and PTSD, making it a potential therapeutic target. A schematic summary has been demonstrated in Fig. 3.
Fig. 3.
Demonstrates a full graphical summary. (A) shows the Cross-talk Between NO, with CO and H2S, (B) shows NO-Mediated Epigenetic Modifications in Memory Consolidation, (C) The biochemical steps for NO synthesis and the important molecular factors required for its production, (D) nitric oxide retrograde messenger, (E) nitric oxide modulation of s-nitrosylation, (F) nitric oxide modulation of CaMKs, (G) nitric oxide regulation of synaptic proteins, (H) nitric oxide impact on AMPAR, (I) nitric oxide role in Alzheimer’s, (J) nitric oxide role in Parkinson’s, K. nitric oxide role in depression, L. nitric oxide role in schizophrenia, M. nitric oxide role in bipolar disorder, N. nitric oxide role in PTSD
Challenges and future directions
Our review highlights several critical gaps in the current understanding of the precise molecular pathway involving NO dysregulation and memory-related disorders. Future research might employ single-cell RNA sequencing to map NO-related gene expression changes during memory consolidation in specific regions of the brain. Furthermore, CRISPR-based genome editing could help identifying the significance of certain nNOS-associated genes in synaptic plasticity. Future studies to monitor NO metabolites in cerebrospinal fluid could aid in the identification of prognostic biomarkers for neurological disorders. Addressing these issues can help in bridging the current gaps related to the role of NO in memory and cognition and enhance the scientific understanding and clinical management of memory-related disorders involved nitric oxide dysregulation and to bring Moreover, advanced imaging techniques should be explored to study the spatial and temporal dynamics of NO in memory consolidation, further advancing this field of research.
Acknowledgements
The publication of this article is funded by Qatar National Library.
Abbreviations
- NO
Nitric oxide
- STM
Short-term memory
- LTM
Long-term memory
- NOS
Nitric oxide synthase
- CAT-1
The cationic amino acid transporter
- eNOS
Endothelial NOS
- nNOS
Neuron NOS
- iNOS
Inflammatory NOS
- BH4
Tetrahydrobiopterin
- FAD
Flavin Adenine Dinucleotide
- FMN
Flavin Mononucleotide
- LTP
Long term potentiation
- NMDA
N-methyl-D-aspartate
- sGC
Soluble guanylate cyclase
- CaMK
Calcium calmodulin-dependent kinase
- Arc
Cytoskeleton-associated protein
- BDNF
Brain-derived neurotrophic factor
- NO-cGMP-PKG
Cyclic guanosine monophosphate-protein kinase G
- MWM
Morris-water maze
- AD
Alzheimer’s disease
- PD
Parkinson’s disease
Author contributions
Conceptualization: AA and RMZ. Study design, data collection, data analysis and Writing-Original draft preparation: ZIB, EAE, RA and AA. Writing- Reviewing and Editing the final manuscript: AA and RMZ. All authors reviewed, contributed, and approved the final manuscript version.
Funding
Open Access funding provided by the Qatar National Library.
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review of all sites.
Informed consent
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Zlotnik G, Vansintjan A (2019) Memory: an extended definition. Front Psychol 10:2523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lipton SA, Choi YB, Pan ZH, Lei SZ, Chen HS, Sucher NJ, Loscalzo J, Singel DJ, Stamler JS (1993) A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds. Nature 364(6438):626–632 [DOI] [PubMed] [Google Scholar]
- 3.Sherwood L (2015) Human physiology: from cells to systems, 9th edn. Cengage Learning
- 4.Malmberg KJ, Raaijmakers JGW, Shiffrin RM (2019) 50 years of research sparked by Atkinson and Shiffrin (1968). Mem Cognit 47(4):561–574 [DOI] [PubMed] [Google Scholar]
- 5.Cowan, N. (2008). Learning and memory: A comprehensive reference (1st ed.). In J. H. Byrne (Ed.), Texas: Academic Press.
- 6.Noushad B, Khurshid F (2019) Facilitating student learning: an instructional design perspective for health professions educators. Res Dev Med Educ 8(2):69–74 [Google Scholar]
- 7.Nursey J, Phelps AJ (2016) In: Fink G (ed) Stress: concepts, cognition, emotion, and Behavior. Elsevier, pp 169–176. 10.1016/C2013-0-12842-5.
- 8.Brewin CR (2014) Episodic memory, perceptual memory, and their interaction: foundations for a theory of posttraumatic stress disorder. Psychol Bull 140(1):69–97 [DOI] [PubMed] [Google Scholar]
- 9.Brickman AM, Stern Y (2009) Encyclopedia of Neuroscience. Elsevier Ltd.
- 10.H.M.A.P.S.a.i.C PRC, Atkinson, Shiffrin RM (1968) pp. 89–195. 10.1016/S0079-7421(08)60422-3
- 11.Almaraz-Espinoza A, Grider MH (2024) Physiology, Long Term Memory [PubMed]
- 12.Smith C, Peters KR (2011) Sleep, memory, and molecular neurobiology. Handb Clin Neurol 98:259–272 [DOI] [PubMed] [Google Scholar]
- 13.Paul V, Ekambaram P (2011) Involvement of nitric oxide in learning & memory processes. Indian J Med Res 133(5):471–478 [PMC free article] [PubMed] [Google Scholar]
- 14.Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield DA, Stella AM (2007) Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci 8(10):766–775 [DOI] [PubMed] [Google Scholar]
- 15.Garthwaite J, Boulton CL (1995) Nitric oxide signaling in the central nervous system. Annu Rev Physiol 57:683–706 [DOI] [PubMed] [Google Scholar]
- 16.Ghasemi M (2019) Nitric oxide: antidepressant mechanisms and inflammation. Adv Pharmacol 86:121–152 [DOI] [PubMed] [Google Scholar]
- 17.Gorren AC, Mayer B (2002) Tetrahydrobiopterin in nitric oxide synthesis: a novel biological role for pteridines. Curr Drug Metab 3(2):133–157 [DOI] [PubMed] [Google Scholar]
- 18.Feng Y, Feng Y, Gu L, Liu P, Cao J, Zhang S (2021) The critical role of tetrahydrobiopterin (BH4) metabolism in modulating radiosensitivity: BH4/NOS Axis as an Angel or a Devil. Front Oncol 11:720632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M (2023) Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol 97(10):2499–2574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Squire LR, Genzel L, Wixted JT, Morris RG (2015) Memory consolidation. Cold Spring Harb Perspect Biol 7(8):a021766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Goto A (2022) Synaptic plasticity during systems memory consolidation. Neurosci Res 183:1–6 [DOI] [PubMed] [Google Scholar]
- 22.Bon CL, Garthwaite J (2003) On the role of nitric oxide in hippocampal long-term potentiation. J Neurosci 23(5):1941–1948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hardingham N, Dachtler J, Fox K (2013) The role of nitric oxide in pre-synaptic plasticity and homeostasis. Front Cell Neurosci 7:190 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Malenka RC, Lancaster B, Zucker RS (1992) Temporal limits on the rise in postsynaptic calcium required for the induction of long-term potentiation. Neuron 9(1):121–128 [DOI] [PubMed] [Google Scholar]
- 25.Wittenborn EC, Marletta MA (2021) Structural perspectives on the mechanism of Soluble Guanylate Cyclase activation. Int J Mol Sci 22(11) [DOI] [PMC free article] [PubMed]
- 26.Li Y, Xu X, Qi G, Cui D, Huang C, Sui X, Li G, Fan Q (2023) Mechanisms of autophagy function and regulation in plant growth, development, and response to abiotic stress. Crop J 11(6):1611–1625 [Google Scholar]
- 27.Nakamura T, Lipton SA (2010) Preventing Ca2+-mediated nitrosative stress in neurodegenerative diseases: possible pharmacological strategies. Cell Calcium 47(2):190–197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Nakamura T, Lipton SA (2007) S-Nitrosylation and uncompetitive/fast off-rate (UFO) drug therapy in neurodegenerative disorders of protein misfolding. Cell Death Differ 14(7):1305–1314 [DOI] [PubMed] [Google Scholar]
- 29.Iova OM, Marin GE, Lazar I, Stanescu I, Dogaru G, Nicula CA, Bulboaca AE (2023) Nitric Oxide/Nitric oxide synthase system in the pathogenesis of neurodegenerative Disorders-An overview. Antioxid (Basel) 12(3) [DOI] [PMC free article] [PubMed]
- 30.Santos AI, Martinez-Ruiz A, Araujo IM (2015) S-nitrosation and neuronal plasticity. Br J Pharmacol 172(6):1468–1478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Forrester MT, Foster MW, Benhar M, Stamler JS (2009) Detection of protein S-nitrosylation with the biotin-switch technique. Free Radic Biol Med 46(2):119–126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Song T, Hatano N, Kambe T, Miyamoto Y, Ihara H, Yamamoto H, Sugimoto K, Kume K, Yamaguchi F, Tokuda M, Watanabe Y (2008) Nitric oxide-mediated modulation of calcium/calmodulin-dependent protein kinase II. Biochem J 412(2):223–231 [DOI] [PubMed] [Google Scholar]
- 33.Soderling TR, Chang B, Brickey D (2001) Cellular signaling through multifunctional Ca2+/calmodulin-dependent protein kinase II. J Biol Chem 276(6):3719–3722 [DOI] [PubMed] [Google Scholar]
- 34.Araki S, Osuka K, Takata T, Tsuchiya Y, Watanabe Y (2020) Coordination between Calcium/Calmodulin-Dependent protein kinase II and neuronal nitric oxide synthase in neurons. Int J Mol Sci 21(21) [DOI] [PMC free article] [PubMed]
- 35.Costa C, Tozzi A, Siliquini S, Galletti F, Cardaioli G, Tantucci M, Pisani F, Calabresi P (2011) A critical role of NO/cGMP/PKG dependent pathway in hippocampal post-ischemic LTP: modulation by zonisamide. Neurobiol Dis 44(2):185–191 [DOI] [PubMed] [Google Scholar]
- 36.Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV (2022) Role of ca(2+)/Calmodulin-Dependent protein kinase type II in mediating function and dysfunction at glutamatergic synapses. Front Mol Neurosci 15:855752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lu YF, Kandel ER, Hawkins RD (1999) Nitric oxide signaling contributes to late-phase LTP and CREB phosphorylation in the hippocampus. J Neurosci 19(23):10250–10261 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kida S (2012) A functional role for CREB as a positive Regulator of memory formation and LTP. Exp Neurobiol 21(4):136–140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Finkbeiner S, Tavazoie SF, Maloratsky A, Jacobs KM, Harris KM, Greenberg ME (1997) CREB: a major mediator of neuronal neurotrophin responses. Neuron 19(5):1031–1047 [DOI] [PubMed] [Google Scholar]
- 40.Snow WM, Pahlavan PS, Djordjevic J, McAllister D, Platt EE, Alashmali S, Bernstein MJ, Suh M, Albensi BC (2015) Morris Water Maze Training in mice elevates hippocampal levels of transcription factors nuclear factor (erythroid-derived 2)-like 2 and nuclear factor Kappa B p65. Front Mol Neurosci 8:70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bencsik N, Oueslati Morales CO, Hausser A, Schlett K (2023) Endocytosis of AMPA receptors: role in neurological conditions. Prog Mol Biol Transl Sci 196:59–97 [DOI] [PubMed] [Google Scholar]
- 42.Ivanova VO, Balaban PM, Bal NV (2020) Modulation of AMPA receptors by nitric oxide in nerve cells. Int J Mol Sci 21(3) [DOI] [PMC free article] [PubMed]
- 43.Ota KT, Pierre VJ, Ploski JE, Queen K, Schafe GE (2008) The NO-cGMP-PKG signaling pathway regulates synaptic plasticity and fear memory consolidation in the lateral amygdala via activation of ERK/MAP kinase. Learn Mem 15(10):792–805 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Schafe GE, Bauer EP, Rosis S, Farb CR, Rodrigues SM, LeDoux JE (2005) Memory consolidation of pavlovian fear conditioning requires nitric oxide signaling in the lateral amygdala. Eur J Neurosci 22(1):201–211 [DOI] [PubMed] [Google Scholar]
- 45.Rodkin S, Nwosu C, Sannikov A, Tyurin A, Chulkov VS, Raevskaya M, Ermakov A, Kirichenko E, Gasanov M (2023) The role of gasotransmitter-dependent signaling mechanisms in apoptotic cell death in Cardiovascular, rheumatic, kidney, and neurodegenerative diseases and Mental disorders. Int J Mol Sci 24(7) [DOI] [PMC free article] [PubMed]
- 46.Mir JM, Maurya RC (2018) A gentle introduction to gasotransmitters with special reference to nitric oxide: biological and chemical implications. Rev Inorg Chem 38(4):193–220 [Google Scholar]
- 47.Huang EP (1997) Synaptic plasticity: a role for nitric oxide in LTP. Curr Biol 7(3):R141–R143 [DOI] [PubMed] [Google Scholar]
- 48.Zhuo M, Hu Y, Schultz C, Kandel ER, Hawkins RD (1994) Role of guanylyl cyclase and cGMP-dependent protein kinase in long-term potentiation. Nature 368(6472):635–639 [DOI] [PubMed] [Google Scholar]
- 49.Kirchner L, Weitzdoerfer R, Hoeger H, Url A, Schmidt P, Engelmann M, Villar SR, Fountoulakis M, Lubec G, Lubec B (2004) Impaired cognitive performance in neuronal nitric oxide synthase knockout mice is associated with hippocampal protein derangements. Nitric Oxide 11(4):316–330 [DOI] [PubMed] [Google Scholar]
- 50.Chueh AC, Tse JW, Togel L, Mariadason JM (2015) Mechanisms of histone deacetylase inhibitor-regulated gene expression in Cancer cells. Antioxid Redox Signal 23(1):66–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Itzhak Y, Anderson KL, Kelley JB, Petkov M (2012) Histone acetylation rescues contextual fear conditioning in nNOS KO mice and accelerates extinction of cued fear conditioning in wild type mice. Neurobiol Learn Mem 97(4):409–417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zovkic IB, Guzman-Karlsson MC, Sweatt JD (2013) Epigenetic regulation of memory formation and maintenance. Learn Mem 20(2):61–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Asiimwe N, Yeo SG, Kim MS, Jung J, Jeong NY (2016) Nitric Oxide: Exploring the Contextual Link with Alzheimer’s Disease, Oxid Med Cell Longev (2016) 7205747 [DOI] [PMC free article] [PubMed]
- 54.Azargoonjahromi A (2023) Dual role of nitric oxide in Alzheimer’s disease. Nitric Oxide 134-135, 23–37. [DOI] [PubMed]
- 55.Stykel MG, Ryan SD (2022) Nitrosative stress in Parkinson’s disease. NPJ Parkinsons Dis 8(1):104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Nasyrova RF, Ivashchenko DV, Ivanov MV, Neznanov NG (2015) Role of nitric oxide and related molecules in schizophrenia pathogenesis: biochemical, genetic and clinical aspects. Front Physiol 6:139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Somani A, Singh AK, Gupta B, Nagarkoti S, Dalal PK, Dikshit M (2022) Oxidative and nitrosative stress in major depressive disorder: a Case Control Study. Brain Sci 12(2) [DOI] [PMC free article] [PubMed]
- 58.Massey N, Vasanthi SS, Samidurai M, Gage M, Rao N, Meyer C, Thippeswamy T (2023) 1400 W, a selective inducible nitric oxide synthase inhibitor, mitigates early neuroinflammation and nitrooxidative stress in diisopropylfluorophosphate-induced short-term neurotoxicity rat model. Front Mol Neurosci 16:1125934 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cuadrado-Tejedor M, Hervias I, Ricobaraza A, Puerta E, Perez-Roldan JM, Garcia-Barroso C, Franco R, Aguirre N, Garcia-Osta A (2011) Sildenafil restores cognitive function without affecting beta-amyloid burden in a mouse model of Alzheimer’s disease. Br J Pharmacol 164(8):2029–2041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dean O, Giorlando F, Berk M (2011) N-acetylcysteine in psychiatry: current therapeutic evidence and potential mechanisms of action. J Psychiatry Neurosci 36(2):78–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Soria Lopez JA, Gonzalez HM, Leger GC (2019) Alzheimer’s disease. Handb Clin Neurol 167:231–255 [DOI] [PubMed] [Google Scholar]
- 62.Venturelli M, Pedrinolla A, Boscolo Galazzo I, Fonte C, Smania N, Tamburin S, Muti E, Crispoltoni L, Stabile A, Pistilli A, Rende M, Pizzini FB, Schena F (2018) Impact of nitric oxide bioavailability on the progressive cerebral and peripheral circulatory impairments during aging and Alzheimer’s Disease. Front Physiol 9:169 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Huie RE, Padmaja S (1993) The reaction of no with superoxide. Free Radic Res Commun 18(4):195–199 [DOI] [PubMed] [Google Scholar]
- 64.Pacher P, Beckman JS, Liaudet L (2007) Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87(1):315–424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tonnies E, Trushina E, Stress O (2017) Synaptic dysfunction, and Alzheimer’s Disease. J Alzheimers Dis 57(4):1105–1121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lazzaro M, Bettegazzi B, Barbariga M, Codazzi F, Zacchetti D, Alessio M (2014) Ceruloplasmin potentiates nitric oxide synthase activity and cytokine secretion in activated microglia. J Neuroinflammation 11:164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O’Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T (2000) Inflammation and Alzheimer’s disease. Neurobiol Aging 21(3):383–421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Liu J, Chang L, Song Y, Li H, Wu Y (2019) The role of NMDA receptors in Alzheimer’s Disease. Front Neurosci 13:43 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dawson VL, Dawson TM (1998) Nitric oxide in neurodegeneration. Prog Brain Res 118:215–229 [DOI] [PubMed] [Google Scholar]
- 70.Zhang L, Dawson VL, Dawson TM (2006) Role of nitric oxide in Parkinson’s disease. Pharmacol Ther 109(1–2):33–41 [DOI] [PubMed] [Google Scholar]
- 71.Hirsch EC, Jenner P, Przedborski S (2013) Pathogenesis of Parkinson’s disease. Mov Disord 28(1):24–30 [DOI] [PubMed] [Google Scholar]
- 72.Vaidya B, Sharma SS (2023) Nitric oxide in Parkinson’s Disease: insights into research and therapeutics. In: Ray A, Gulati K (eds) Nitric oxide: from research to therapeutics. Springer International Publishing, Cham, pp 327–347 [Google Scholar]
- 73.Jauhar S, Johnstone M, McKenna PJ (2022) Schizophrenia Lancet 399(10323):473–486 [DOI] [PubMed] [Google Scholar]
- 74.Ben-Shachar D, Laifenfeld D (2004) Mitochondria, synaptic plasticity, and schizophrenia. Int Rev Neurobiol 59:273–296 [DOI] [PubMed] [Google Scholar]
- 75.Stampanoni Bassi M, Iezzi E, Gilio L, Centonze D, Buttari F (2019) Synaptic plasticity shapes Brain Connectivity: implications for Network Topology. Int J Mol Sci 20(24) [DOI] [PMC free article] [PubMed]
- 76.Clos M, Bunzeck N, Sommer T (2019) Dopamine is a double-edged sword: dopaminergic modulation enhances memory retrieval performance but impairs metacognition. Neuropsychopharmacology 44(3):555–563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Dhir A, Kulkarni SK (2011) Nitric oxide and major depression. Nitric Oxide 24(3):125–131 [DOI] [PubMed] [Google Scholar]
- 78.Suzuki E, Yagi G, Nakaki T, Kanba S, Asai M (2001) Elevated plasma nitrate levels in depressive states. J Affect Disord 63(1–3):221–224 [DOI] [PubMed] [Google Scholar]
- 79.Zhou QG, Hu Y, Hua Y, Hu M, Luo CX, Han X, Zhu XJ, Wang B, Xu JS, Zhu DY (2007) Neuronal nitric oxide synthase contributes to chronic stress-induced depression by suppressing hippocampal neurogenesis. J Neurochem 103(5):1843–1854 [DOI] [PubMed] [Google Scholar]
- 80.Wang W, Zhou T, Jia R, Zhang H, Zhang Y, Wang C, Dong Y, Wang J, Sheng L, Wu H, Chen G, Xue W (2019) NMDA receptors and L-arginine/nitric oxide/cyclic guanosine monophosphate pathway contribute to the antidepressant-like effect of Yueju pill in mice. Biosci Rep 39(9) [DOI] [PMC free article] [PubMed]
- 81.Fanet H, Capuron L, Castanon N, Calon F, Vancassel S (2021) Tetrahydrobioterin (BH4) pathway: from metabolism to Neuropsychiatry. Curr Neuropharmacol 19(5):591–609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chanrion B, Mannoury la Cour C, Bertaso F, Lerner-Natoli M, Freissmuth M, Millan MJ, Bockaert J, Marin P (2007) Physical interaction between the serotonin transporter and neuronal nitric oxide synthase underlies reciprocal modulation of their activity. Proc Natl Acad Sci U S A 104(19):8119–8124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Paul V, Ekambaram P (2011) Involvement of nitric oxide in learning & memory processes. Indian J Med Res 133(5):471–478 PMID: 21623030; PMCID: PMC3121276.) [PMC free article] [PubMed] [Google Scholar]
- 84.Tricoire L, Vitalis T (2012) Neuronal nitric oxide synthase expressing neurons: a journey from birth to neuronal circuits. Front Neural Circuits 6:82 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ustundag MF, Ozcan H, Gencer AG, Yilmaz ED, Ugur K, Oral E, Bilici M (2020) Nitric oxide, asymmetric dimethylarginine, symmetric dimethylarginine and L-arginine levels in psychotic exacerbation of schizophrenia and bipolar disorder manic episode. Saudi Med J 41(1):38–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Morris G, Reiche EMV, Murru A, Carvalho AF, Maes M, Berk M, Puri BK (2018) Multiple Immune-Inflammatory and oxidative and nitrosative stress pathways explain the frequent Presence of Depression in multiple sclerosis. Mol Neurobiol 55(8):6282–6306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Fronza MG, Ferreira BF, Pavan-Silva I, Guimaraes FS, Lisboa SF (2023) NO time in fear response: possible implication of nitric-oxide-related mechanisms in PTSD. Molecules 29(1) [DOI] [PMC free article] [PubMed]
- 88.Lawrence S, Scofield RH (2024) Post traumatic stress disorder associated hypothalamic-pituitary-adrenal axis dysregulation and physical illness. Brain Behav Immun Health 41:100849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sadeghi MA, Hemmati S, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Chamanara M (2022) Targeting neuronal nitric oxide synthase and the nitrergic system in post-traumatic stress disorder. Psychopharmacology 239(10):3057–3082 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.



