Next Article in Journal
Asthma and COPD Beyond the Airways: Exploring Neurocognitive Links Through NF-κB Subunits c-Rel and p65
Previous Article in Journal
Hyperarousal, Dissociation, Emotion Dysregulation and Re-Experiencing—Towards Understanding Molecular Aspects of PTSD Symptoms
Previous Article in Special Issue
The Role of microRNAs in Epigenetic Regulation of Signaling Pathways in Neurological Pathologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Serotonergic Regulation in Alzheimer’s Disease

by
Lyudmila P. Dolgacheva
1,
Valery P. Zinchenko
1,
Alexander D. Nadeev
1 and
Nikolay V. Goncharov
2,*
1
Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia
2
Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(11), 5218; https://doi.org/10.3390/ijms26115218
Submission received: 28 March 2025 / Revised: 18 May 2025 / Accepted: 27 May 2025 / Published: 29 May 2025

Abstract

Serotonin (5-HT) is a neurotransmitter that also plays an important role in the regulation of vascular tone and angiogenesis. This review focuses on the involvement of the 5-HT system in pathological processes leading to the development of Alzheimer’s disease (AD). There is evidence that damage or dysfunction of the 5-HT system contributes to the development of AD, and different subtypes of 5-HT receptors are a potential target for the treatment of AD. A link has been established between AD, depression, stress, and 5-HT deficiency in the brain. There are new data on the role of circadian rhythms in modulating stress, depression, and the 5-HT system; amyloid β (Aβ) plaque clearance; and AD progression. Circadian disruption inhibits Aβ plaque clearance and modulates AD progression. The properties and functions of 5-HT, its receptors, and serotonergic neurons are presented. Special attention is paid to the central role of 5-HT in brain development, including neurite outgrowth, regulation of somatic morphology, motility, synaptogenesis, control of dendritic spine shape and density, neuronal plasticity determining its role in network regeneration, and changes in innervation after brain damage. The results of different studies indicate that the interaction of amyloid β oligomers (AβO) with mitochondria is a sufficient trigger for AD-related neurodegeneration. The action of 5-HT leads to an improvement in mitochondrial quality and the restoration of brain areas after traumatic brain injury, chronic stress, or developmental disorders in AD. The role of a healthy lifestyle and drugs acting on serotonin receptors in the prevention and treatment of AD is discussed.

1. Introduction

Recent studies increasingly point to the involvement of serotoninergic neurons in the pathological processes underlying AD. Damage to these neurons is associated with the deterioration of cognitive processes not only in aging and AD, but also in other mental disorders, including schizophrenia, stress, and depression [1,2,3].
The 5-HT system is multifaceted, affecting general mechanisms of AD and other neuropathologies, as well as specific mechanisms unique to AD. For example, the 5-HT system is involved in the control of neuroinflammation, oxidative stress, and mitochondrial dysfunction, which contribute to the development of a wide range of neuropathologies [4,5,6,7,8].
On the other hand, a growing body of data demonstrates the involvement of the 5-HT system in the mechanisms of formation of insoluble aggregates of Aβ and (hyper)phosphorylated tau protein (pTau), which are the main histopathological signs of Alzheimer’s disease [9]. This disorder is characterized by the accumulation of two abnormally folded proteins: β-amyloid (Aβ), forming extracellular senile plaques, and pTau, forming intracellular tangles. Different subtypes of 5-HT receptors and the intracellular signaling cascades they trigger are involved in various neuropathologies. Therefore, agonists and antagonists of different 5-HT receptors are being actively studied both as antidepressants and antipsychotics for the treatment of behavioral symptoms of different neuropathologies [10], and as drugs that affect specific causes of AD, namely the accumulation of Aβ and pTau [11]. Dysfunction of the 5-HT system occurs in the development of AD [1], but the mechanism of dysfunction is not fully understood. The number of 5-HT neurons in the dorsal raphe nuclei, the levels of 5-HT and its metabolite 5-hydroxyindoleacetic acid, and the binding strength for 5-HT in the cortex and hippocampus are significantly reduced in AD [12].
Selective 5-HT reuptake inhibitors, including escitalopram, citalopram, and fluoxetine, have beneficial effects on both psychiatric symptoms and, to a lesser extent, cognitive impairment in patients with AD [13,14,15]. Serotonin-selective antidepressants significantly improve depressive symptoms and daily functioning in patients with AD and are used to treat aggressive behavior. By modulating both the proliferation and survival of newly formed cells, serotonin is a key regulator of adult neurogenesis [16].

2. Stress, Depression, and the Development of AD

Alzheimer’s disease is a chronic neurodegenerative disorder that occurs as a result of the combined action of a number of factors: genetic disorders, aging processes, and accumulation of protein aggregates. AD causes progressive dysfunction and death of neurons in the cerebral cortex and certain subcortical areas associated with cognitive abilities and memory [17]. In AD, neurons associated with memory in the cortex and hippocampus are initially destroyed [18]. Later, areas of the cerebral cortex responsible for spatial navigation, language, thinking, and social behavior are affected.
AD occurs in two forms: familial (hereditary) and sporadic. In the hereditary form, genetic mutations are observed in the presenilin 1 and 2 (PSEN1 and PSEN2) and amyloid precursor protein (APP) genes. Mutations in these genes increase Aβ production, raise the Aβ42/Aβ40 ratio, and promote plaque formation [19]. The combined effect of unhealthy lifestyle, environment, and genetic factors causes the sporadic form of the disease.
The most common manifestations of AD are short-term memory impairment and impaired expressive speech. Morphologically, AD is characterized by the formation of extracellular amyloid plaques, consisting mainly of fibrillar Aβ, as well as intracellular neurofibrillary tangles (NFTs), consisting of pTau. These two toxic protein compounds, Aβ and pTau, come into play at different time stages of the disease [20,21]. Aβ accumulation has been shown to be a marker of early Alzheimer’s disease, and pTau is an indicator of late stages of the disease [22,23]. Both Aβ and pTau affect the electron transport chain (ETC), which affects energy production [24,25], and also induce mitophagy, which results in excessive loss of mitochondria [26,27].
The Aβ protein is a central component of extracellular amyloid plaques in AD. Aβ causes mitochondrial dysfunction by inhibiting mitochondrial transmembrane translocase, interacting with cyclophilin D to interfere with energy production, and promoting opening of the mitochondrial highly permeable pore (mPTP) [25,28]. Aβ peptides are produced by the enzymatic cleavage of APP [29]. APP is a transmembrane glycoprotein expressed largely within the central nervous system (CNS). It is known that APP plays a significant role in neural growth and maturation during brain development. Full-length APP is incorporated not only into the plasma membrane, but also into the membranes of intracellular organelles, where it can be converted via the non-amyloidogenic or amyloidogenic pathway. Under physiological conditions (non-amyloidogenic pathways), APP is primarily first cleaved by α-secretase, producing a secreted form of APP (sAPPα) and a membrane-bound C-terminal fragment of 83 amino acids (CTF, C8), preventing Aβ production [30]. The non-amyloidogenic pathway plays a significant role in maintaining neuronal homeostasis, in signaling and intracellular transport, and is also involved in synaptogenesis and synaptic plasticity [31]. The non-amyloidogenic pathway is the most predominant proteolytic processing of the APP in healthy brain [32]. The secreted form of sAPPα exerts beneficial physiological, biochemical, and behavioral effects by counteracting the detrimental effects of neurotoxic agents. It has been shown to stimulate neurite outgrowth in neurons, promote clearance of Aβ from damaged microglia by restoring mitochondrial function, and reverse age-related declines in neural progenitor cell proliferation [33,34,35].
Amyloidogenic proteolytic processing of APP on the cell surface leads to the formation of Aβ [36,37]. Aβ oligomers (AβOs) alter the morphology and density of synapses and disrupt synaptic plasticity. Impaired synaptic function is considered an early and key pathology of AD. The most common Aβ species are 27–43 amino acids in length. Once produced, these short Aβ isoforms are secreted into the extracellular space as monomers via exosomes [38]. Long intracellular Aβ (iAβ) isoforms, such as Aβ45, aggregate in neurons [39,40]. Phosphorylation also causes protein aggregation and insoluble protein formation. iAβ accumulation is associated with the glymphatic system, particularly the paravascular pathway, which plays a key role in the clearance of extracellular Aβ mediated by astroglial aquaporin (AQP4) [19]. The release of Aβ fragments first leads to the spontaneous aggregation of Aβ monomers into soluble AβO, which is promoted by the presence of hydrophobic amino acids. The further aggregation of oligomers results in the formation of insoluble protofibrils [41].
AβO can exert toxic effects by directly interacting with pTau, lipid, and cholesterol components of the cell membrane, leading to the formation of channels and the destruction of membrane integrity and permeability. The latter allows Ca2+ ions to enter the cell, inhibit long-term potentiation (LTP), and induce neuronal death [42,43]. In addition, AβO contributes to other pathological manifestations of AD, including neuroinflammation, oxidative stress, and mitochondrial dysfunction, which also lead to neuronal death. Aβ levels are also modulated by the sleep–wake cycle [44]. A higher production and release of Aβ into the extracellular space has been observed during wakefulness, while enhanced Aβ clearance via the glymphatic system has been documented during sleep [45].
Stress and depression make a significant contribution to the development of AD [46,47]. Symptoms of depression are numerous and varied. They include apathy, feelings of guilt, worthlessness, hopelessness over a long period of time, sleep problems, and inability to concentrate [48,49,50]. Depression is thought to accelerate neurodegenerative processes, as individuals with depression are more likely to develop AD [51]. AβO increases long-term depression (LTD) in the hippocampus. Clinical data indicate that in both pathologies, atrophic changes occur in the same areas of the brain—the hippocampus and prefrontal cortex (PFC). These brain regions play a key role in regulating the stress response. Thus, depression may be an initial sign of neurodegenerative disease and may be considered a risk factor for the further development of AD [47].
Proposing that a deficiency of brain monoamines, including 5-HT, triggers the onset of depression, the monoamine–serotonin hypothesis of depression was proposed in the 1960s [52,53,54]. Acute depletion of tryptophan (a precursor of serotonin) causes the relapse of depression in patients, and the administration of 5-HT antidepressants causes remission [55,56,57]. Selective serotonin reuptake inhibitors (SSRIs) are common protective drugs against LTD [58].
The circadian and stress systems are also closely interconnected. Normally, the stress system is under circadian control and optimizes interactions with the environment [59]. In turn, the stress system can have feedback with the circadian system and provide information about time to the entire organism [59]. People with depression are more likely to have changes in their circadian rhythms, such as sleep–wake cycles, body temperature, neurotransmitter levels, and hormones [60]. In mammals, the circadian system is controlled by a central pacemaker known as the suprachiasmatic nucleus (SCN) of the hypothalamus [61]. The circadian system not only indirectly but also directly controls the development of AD. In experimental models of AD in mice, intact circadian rhythms promoted the removal of Aβ plaques, whereas the disruption of circadian rhythms inhibited their removal [62,63].
At the same time, the circadian system is closely linked to the stress-sensitive serotonin system [64]. There is a bidirectional relationship between 5-HT and the circadian system. Serotonergic antidepressant drugs treat the consequences of circadian rhythm disruption and, conversely, the 5-HT system is under circadian control. At the genetic level, key signaling molecules of the serotonin signaling network, such as the Na+/Cl-dependent serotonin transporter (SERT) and the 5-HT1B, 5-HT7, and 5-HT2C receptors, are expressed in the biological clock core SCN [65,66,67,68], where they mediate the effects of serotonin on rhythms. Early-morning light exposure [69], daytime exercise [70], and time-restricted diets [71,72] are all helpful in combating symptoms of depression, protecting against memory loss, and promoting longevity.
The activity of the glymphatic system, which regulates amyloid clearance from the brain via the perivascular space surrounding blood vessels, is also regulated by circadian rhythms. Dysfunction of the glymphatic system also plays a vital role in the severity of AD. The activity of this system is higher during sleep and lower during wakefulness [73,74]. In addition to local protein degradation mechanisms, plaque clearance via the glymphatic system is inhibited in AD when circadian rhythms are dysregulated. Acute sleep deprivation increases Aβ accumulation in humans [75].

3. AβO, 5-HT, and Mitochondrial Dysfunction

Mitochondria are essential organelles that provide neuronal functioning through their part in energy production, calcium transport, maintenance of redox balance, and programmed cell death [76]. Mitochondria are one of the main sources of reactive oxygen species (ROS). High levels of ROS can impair cell integrity through oxidation of lipids, proteins, and DNA. Many neurodegenerative diseases are accompanied by mitochondrial dysfunction [77,78].
AβO is considered to be the main cause of toxic effects in the AD brain [79,80]. The physical damage is partially attributable to the pore-forming effect of AβO [81]. AβO can incorporate directly into lipid bilayers, changing the structure of the cell membrane and thereby altering its permeability [82]. The barrel-like structure of AβO facilitates the formation of ion channels within the cell membrane, thereby enabling Ca2+ entry into the cell. It has been established that aggregated Aβ42 has the capacity to form ion channels, a property that is absent in Aβ40 [81]. It is hypothesized that another potential cause of mechanical damage to neurons is the interaction between AβO and lipid rafts [83].
A substantial number of reports have identified various forms of AβO receptors, as well as the associated signaling proteins. Well-studied AβO receptors include the cellular prion protein (PrPc) [84] and the neuron-specific Na/K-ATPase α3 subunit (NKAα3), which can promote neurodegeneration through presynaptic calcium overload [85]. PrPc binds to AβO, reducing the NMDA receptors’ (NMDARs’) density at synapses and leading to a loss of dendritic spines [86]. PrPc also forms complexes with AβO, activating metabotropic glutamate receptor 5 and inducing Ca2+ influx. Several other receptors have also been reported to bind directly or indirectly to AβO, thereby affecting neuronal survival.
In addition, several receptors have been reported to bind to AβO, allowing them to enter neurons in an endocytic manner. Aβ accumulates in lysosomal vesicles, which have a low internal pH. The acidic environment within endosomes and lysosomes serves as a preferential site for AβO formation [87]. The rate of assembly of AβO increases 8000-fold when they migrate from the neutral extracellular medium to the low-pH lysosomal environment [88]. Selective antibodies to AβO prevent neuropathology in a mouse model of AD and restore cognitive impairment in mice for at least 40 days [80,89,90]. Numerous data indicate that AβO receptors are not only necessary but also sufficient triggers of AD-associated neurodegeneration.
Increasing evidence suggests that AβO impairs mitochondrial function by disrupting mitochondrial dynamics and cristae structure, reducing mitochondrial quantity and quality, causing defects in mDNA, and inhibiting the mitochondrial ETC [91,92,93,94,95,96]. The resulting mitochondrial dysfunction initiates cell death processes.
It is currently known that Aβ accumulation in mitochondria is carried out by translocases of the outer (TOMs) and inner membranes (TIMs) [97,98]. It was reported by Sirk et al. that the rates of entry of two endogenous nuclear-encoded mitochondrial proteins, mortalin (mtHsp70) and TOM20, into mitochondria decreased under conditions of sublethal Aβ42 exposure, suggesting that Aβ42 occupies the TOM complex, thereby impairing its mitochondrial entry [99]. Inhibition of TOM20, TOM70, and TOM40 led to reduced Aβ accumulation in mitochondria. The presence of AβO can lead to an imbalance in mitochondrial dynamics that is manifested by increased mitochondrial fission and fusion, and increased mitochondrial fragmentation [100].
In neurons, AβO induces mitochondrial dysfunction and inhibits mitochondrial autophagy, leading to the release of mitochondrial contents, which act as the top sensors of PANoptosis (apoptosis, necroptosis, and pyroptosis) [100]. ETC damage by AβO is the main cause of decreased oxidative phosphorylation. In addition to Aβ and pTau accumulation, AD pathogenesis in mitochondria is associated with increased mDNA damage, low synaptic ATP, increased oxidative stress, and defective autophagy and mitophagy. Dysfunctional mitochondria accelerate the pathological process by increasing phosphorylation of tau proteins, generating excessive amounts of ROS, and activating pathways that lead to cell death [77]. Serotonin plays an important role in the regulation/restoration of synaptic transmission of neurons damaged in AD by improving mitochondrial function (Figure 1).

4. Metabolism and Functions of Serotonin in the CNS

Serotonergic neurons are highly branched, with axons reaching all areas of the brain. Serotonin regulates many physiological processes in neuropathologies, including neuronal stability, energy homeostasis, circadian rhythm, and neurogenesis [101,102]. By increasing neurogenesis in the hippocampus, 5-HT may exert an antidepressant effect [101,102].
Serotonin is synthesized from the amino acid L-tryptophan and is unable to penetrate the CNS through the blood–brain barrier [103]. L-tryptophan from the gut enters the bloodstream, binds to blood albumin, and is transported across the blood–brain barrier to the CNS by a specific neutral amino acid carrier [104]. 5-HT synthesis in the CNS occurs in serotonergic neurons that are located in two relatively small dorsal and median raphe nuclei (DRN and MRN) of the reticular formation of the brainstem [105]. The synthesis of 5-HT is a two-step process. The first step, catalyzed by the rate-limiting enzyme tryptophan hydroxylase 2 (Tph2), is the oxidation of tryptophan to 5-hydroxytryptophan (5-HTP) [106]. The second step is the conversion of 5-HTP into serotonin (5-hydroxytryptamine, 5-HT) by aromatic L-amino acid decarboxylase. Serotonin is an antioxidant that inhibits the generation of ROS, malondialdehyde and carbonyls, prevents thiol oxidation, reduces the degradation of 2-deoxy-D-ribose, and prevents apoptosis [107]. The important role of 5-HT as a regulator of mitochondrial biogenesis and function has been demonstrated in rodent cortical neurons: 5-HT increased mitochondrial biogenesis, as reflected by increased mtDNA levels and mitochondrial gene expression [108]. This was accompanied by increases in cellular ATP, basal and maximal respiration, and reserve respiratory capacity. These mitochondrial effects play important roles in cell survival, neuronal plasticity, stress adaptation, and the regulation of aging [108].
Serotonin plays two key roles. First, during early development, it acts as a growth factor, regulating the development of the nervous system [109,110]. 5-HT is involved in many aspects of neuronal development, including neurite outgrowth, synaptogenesis, control of dendritic spine shape and density, and the development and function of key stress response systems [111,112,113]. In doing so, the 5-HT system interacts with the brain-derived neurotrophic factor (BDNF), S100beta, and other chemical messengers (Figure 2). As a developmental trophic factor, 5-HT regulates a variety of important processes, such as cell division, survival, differentiation, migration, myelination, synaptogenesis, and dendrite pruning [109,114]. A unique property of the 5-HT system is the ability to restore and re-innervate brain areas after a traumatic brain injury and chronic stress [115,116,117]. By regulating neurite outgrowth and synaptogenesis, 5-HT exerts profound effects on neuronal plasticity in the developing and mature nervous system [118,119]. The resulting changes in early-life control have critical consequences for behavior and mental health throughout life [120]. The disruption of these developmental processes may contribute to CNS disorders.
Second, serotonin acts as a potent neuromodulator in the central nervous system across the entire spectrum of vertebrate and invertebrate eukaryotes. As a neurotransmitter, 5-HT plays a central role in brain development and the regulation of attention, pain, mood and emotion, sleep, and arousal. 5-HT also plays a crucial role in the development and function of key stress response systems (the hypothalamic–pituitary–adrenal (HPA) and locus coeruleus–norepinephrine (LC-NE) systems) [111,121]. Neurons of the 5-HT system modulate a wide range of functions, including perception, mood, appetite, aggression, and anxiety [105]. 5-HT is also associated with cognition, memory, learning, sexuality, attention [122], respiratory stability [123], sleep–wake cycles, and circadian rhythms [124]. The serotonergic system also has a modulating effect on some hormones and neurotransmitters, such as dopamine, adrenaline and γ-aminobutyric acid (GABA), cortisol, prolactin, acetylcholine, oxytocin, substance P, and vasopressin [125,126,127]. Neural circuits capable of generating rhythmic behavioral actions, such as breathing, are modulated by the activity of serotonin receptors [128]. The anterior cingulate cortex (ACC) is a key brain region responsible for processing pain information and is under dense serotonergic innervation. The ACC is responsible for a variety of cognitive functions, including emotional expression, attention, and mood regulation.
Imbalances in serotonergic signaling are observed in many pathophysiological conditions, including attention deficit hyperactivity disorder, anxiety disorders, major depressive disorder (MDD), and AD [129,130]. Aβ deposits at the projection sites of serotonergic neurons may cause the retrograde degeneration of these neurons. Serotonin deficiency may be responsible for the increase in aggressive behavior and depression often observed in patients with AD. In several mouse models of AD-like brain amyloidosis, a decrease in the number of serotonin-producing neurons in the raphe nucleus and a decrease in the density of serotonergic fibers in several brain regions, including the neocortex and hippocampus, have been found [7]. SSRIs are common first-line treatments for major depression [54]. Acute depletion of tryptophan (a precursor of serotonin) causes a relapse of mild depressive symptoms in patients who had remitted with 5-HT antidepressants [55,56,57].
The functional effects of 5-HT are inhibited by the SERT of the neurotransmitter–sodium–symporter family [131]. In 5-HT neurons, SERT is expressed near the sites of serotonin release. By the reuptake of serotonin into presynaptic neurons, SERT limits the magnitude and duration of the 5-HT signal. As a result, SERT is a major target of antidepressants. Binding of inhibitors to the allosteric site of human SERT (hSERT) prevents the dissociation of antidepressants bound at the central site and may enhance the efficacy of such antidepressants, potentially reducing their dosage and side effects. In addition to SERT, other transporters are expressed in the brain that can mediate the diffusion of serotonin and other monoamines across cell membranes. These transporters, which include the organic cation transporters OCT1, OCT2, and OCT3, and the plasma membrane monoamine transporter PMAT, are less specific and can move catecholamines and histamine in addition to serotonin. Compared to SERT, they have a relatively low affinity for serotonin but have a significantly higher capacity [132]. Once released, serotonin exerts potent and diverse effects on neurons and other cells through a large family of 5-HT receptors [133] (Figure 2).

5. Serotonin Receptors

The actions of serotonin on target cells, including glutamate and GABA neurons, are mediated by seven receptor subtypes (5-HT1–7) in the central nervous system (CNS), peripheral nervous system (PNS), and blood vessel cells of the cardiovascular system (CVS) (Table 1). Most of them are G-coupled ones and only 5-HT3 receptors are ion channels permeable to sodium and potassium cations [134,135].
The 5-HT1R and 5-HT5R subtypes couple with the Gi/o protein, leading to the inhibition of adenylyl cyclase (AC) activity and a reduction in cyclic adenosine monophosphate (cAMP) levels [136]. 5-HT2R couples with the Gq/11 protein, stimulating phospholipase C, which leads to elevated levels of inositol triphosphate and calcium ions [137]. The 5-HT4, 5-HT6, and 5-HT7 receptors couple with the Gs protein to enhance AC activity and elevate cAMP levels. Notably, 5-HT4R can bind to both Gs and Gi proteins with a preference for Gs over Gi.
Table 1. Serotonin receptors, localization, agonists, and antagonists relationship to AD. References for general and signaling information [135,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]. Abbreviations for signaling: AC—adenylate cyclase; CAS—crk-associated substrate; DAG—diacylglycerol; eNOS—endothelial NO-synthase; ERK1/2—extracellular signal-regulated kinases 1 and 2; HMGB1—high-mobility group protein 1; IP3—inositol 1,4,5-trisphosphate; mTOR—mammalian target of rapamycin; NOX—NADPH oxidase; P38MAPK—p38 mitogen-activated protein kinase; PDK-1—3-phosphoinositide-dependent protein kinase-1; PIP2—phosphatidylinositol 4,5-bisphosphate; PKA—protein kinase A; PKB—protein kinase B (Akt); PKC—protein kinase C; PLC—phospholipase C; ROCK—rho-associated protein kinase; p70S6K1—ribosomal protein S6 kinase beta-1; ROS—reactive oxygen species. Abbreviations for localization: CNS—central nervous system; CVS—cardiovascular system; GI Tract—gastrointestinal tract; PNS—peripheral nervous system; SMC—smooth muscle cells. Chemical names for the research compounds without trivial names are listed in Appendix A.
Table 1. Serotonin receptors, localization, agonists, and antagonists relationship to AD. References for general and signaling information [135,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]. Abbreviations for signaling: AC—adenylate cyclase; CAS—crk-associated substrate; DAG—diacylglycerol; eNOS—endothelial NO-synthase; ERK1/2—extracellular signal-regulated kinases 1 and 2; HMGB1—high-mobility group protein 1; IP3—inositol 1,4,5-trisphosphate; mTOR—mammalian target of rapamycin; NOX—NADPH oxidase; P38MAPK—p38 mitogen-activated protein kinase; PDK-1—3-phosphoinositide-dependent protein kinase-1; PIP2—phosphatidylinositol 4,5-bisphosphate; PKA—protein kinase A; PKB—protein kinase B (Akt); PKC—protein kinase C; PLC—phospholipase C; ROCK—rho-associated protein kinase; p70S6K1—ribosomal protein S6 kinase beta-1; ROS—reactive oxygen species. Abbreviations for localization: CNS—central nervous system; CVS—cardiovascular system; GI Tract—gastrointestinal tract; PNS—peripheral nervous system; SMC—smooth muscle cells. Chemical names for the research compounds without trivial names are listed in Appendix A.
Receptor
Kd
Subtype
SignalingAgonistsAntagonistsLocalizationAD Related
[References]
CNSCVSOther
5-HT1A
2.65 nM
Metabotropic
Gi/Go—AC—PKA
Gi/Go—PI3K—PKB—eNOS
Gi/Go—P38MAPK—HMGB1
Gi/Go—NOX1/4—ROS—ROCK—ERK1/2
Xaliprofen
Ipsapirone
BP 554
8-OH-DPAT
U92016A
WAY100635
NAN190
++
[11]
5-HT1B
16.01 nM
Metabotropic
Gi/Go—AC—PKACGS 12066B
CP-93129
Ergotamine
Eltoprazine
Triptans (Zolmitriptan Sumatriptan, Eletriptan)
Nonyloxytryptamine
GR-127935
GR55562
Isamoltane
SB236057
NAS-181
++
[156]
5-HT1D
10.05 nM
Metabotropic
Gi/Go—AC—PKAPNU-109291
L-703,664
GR 46611
Ergotamine
Alniditan
Triptans (Zolmitriptan, Sumatriptan, Eletriptan, Frovatriptan, Naratriptan, Almotriptan)
SB272183
LY310762
BRL15572
Cyanopindolol
++
[157]
5-HT1E
7.0 nM
Metabotropic
Gi/Go—AC—PKA5-CT
BRL54443
++
5-HT1F
67.60 nM
Metabotropic
Gq/11—PLC—PIP2—IP3—Ca2+—DAG
Gq/11—NOX—ROS—PI3K—PKB—mTOR—p70S6K1
LY334370
BRL54443
LY344864
BRL-54443
Lasmiditan
+
5-HT1P
Metabotropic
5-Hydroxy-indalpine 5-HTP-DP GI Tract
5-HT2A
970.80 nM
Metabotropic
Gq/11—PLC—PIP2—IP3—Ca2+—DAG
Gq/11—ERK1/2—eNOS
DOB
DOI
α-Methyl-5HT
TCB-2
R-95544
Volinanserin
Sarpogrelate
++GI Tract, Platelets, PNS, SMC
[11]
5-HT2B
11.35 nM
Metabotropic
Gq/11—PLC—PIP2—IP3—Ca2+—DAGBW723C86
DOB
DOI
LY272015
RS127445
++GI Tract, Platelets, PNS, SMC
[158]
5-HT2C
35.58 nM
Metabotropic
Gq/11—PLC—PIP2—IP3—Ca2+—DAGWAY 163909
MK212
1-Methyl-psilocin
DOB
DOI
RS102221
SB242084
++GI Tract, Platelets, PNS, SMC
[159]
5-HT3
190.33 nM
Ionotropic
SR57227
2-Methyl-5HT
Phenylbiguanide
MDL 72222
Tropisetron
Ondansetron
Granisetron
+ GI Tract, PNS
[160]
5-HT4
117.0 nM
Metabotropic
Gs—AC—PKARS67506
BIMU1
BIMU8
RS67333
Zacopride
GR113808
RS100235
SB204070
+ GI Tract, PNS
[161]
5-HT5A
Metabotropic
Gi/Go—AC—PKA SB 699551+
[162]
5-HT5B
Metabotropic
Rodent CNS
5-HT6
116.53 нM
Metabotropic
Gs—AC-PKA
Fyn—ras-MEK-ERK1/2
WAY 181,187
EMD 386088
Ro 04-6790
SB 399885
SB271046
+
[163]
5-HT7(a-d)
3.65 нM
Metabotropic
Gs—AC—PKALP12
LP44
AS-19
5-CT
LP211
SB-269970
SB-258719
++GI Tract

[164]

5.1. 5-HT1 Receptors

The 5-HT1 receptor family includes 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E, 5-HT1F, and 5-HT1P receptors that are associated with the Gi/Go protein, the αi subunit of which inhibits AC activity and reduces cAMP levels. Most 5-HT1 receptors are postsynaptic, with the exception of 5-HT1A and 5-HT1B, which are mainly presynaptic and modulate serotonin release. In serotonin neurons, 5-HT1A receptors are located in the somatodendritic compartment [165]. In terminal areas of serotoninergic innervation, such as the hippocampus, the effect of 5-HT1A receptors is realized through both αi and βγ. However, in the dorsal raphe nucleus, 5-HT1A-receptors are associated only with βγ-dependent opening of potassium channels. Through Gβγ subunits, 5-HT1A activation can induce the opening of inwardly rectifying potassium channels (GIRKs), which causes neuronal hyperpolarization [166,167,168]. It has been suggested that 5-HT1A may also regulate not only GIRKs but also small-conductance Ca2+-activated potassium (SK) channels [169]. Most serotonergic neurons have a prominent medium-duration afterhyperpolarization (mAHP), which plays a critical role in setting the firing frequency by delaying the occurrence of the next action potential [170]. This mAHP is due to the opening of SK channels. 5-HT enhances mitochondrial axonal transport in hippocampal neurons via the 5-HT1A receptor [171]. Administration of a 5-HT1A receptor antagonist inhibits mitochondrial movement.
Thus, the ultimate effect of 5-HT1A activation is a decrease in neuronal excitation rate and inhibition of protein kinase A. Activation of presynaptic 5-HT1A receptors by 5-HT provides negative feedback and is considered a key mechanism of autoregulation of the brain 5-HT system [172,173]. Agonists of 5-HT1A, 5-HT1B, and 5-HT1D receptors are being actively tested as therapeutic agents for AD [11,156,157].

5.2. 5-HT2 Receptors

The 5-HT2 receptor family includes the 5-HT2A, 5-HT2B, and 5-HT2C receptors, the 5-HT2A receptors being the most studied. This type of 5-HT receptor is conjugated to the Gq protein that stimulates PLC. PLC hydrolyzes the phospholipid PIP2 to IP3 and DAG, initiating Ca2+ ion elevation and PKC activation [137]. The activation of 5-HT2A receptors mediates neuronal depolarization as a result of the closing of potassium channels (probably due to a decrease in PIP2, an agonist of these channels). The 5-HT2A receptor is the main subtype of serotonin receptors mediating its excitatory effect and increase in neuronal excitability [174,175]. 5-HT2A receptors are localized predominantly on the plasma membrane. However, a number of studies have provided evidence supporting their intracellular localization, for example, under the action of β-arrestin [174,176,177]. The activation of the 5-HT2A-β-arrestin signaling pathway has been shown to initiate an antipsychotic-like behavioral profile [178]. Furthermore, it has been suggested that the association of intracellular 5-HT2A receptors with the microtubule-associated cytoskeletal protein MAP1A may be involved in intraneuronal signaling processes involved in cytoskeletal reorganization [179]. Serotonin regulation of mitochondrial biogenesis and function in rodent cortical neurons is mediated through the 5-HT2A receptor and master modulators of mitochondrial biogenesis, SIRT1 and PGC-1α [108]. The effects of 5-HT on mtDNA, gene expression, ATP levels, and oxidative phosphorylation were observed upon the stimulation of the 5-HT2A receptor of cortical neurons and utilized signaling pathways involving PLC and MAPK, but not PI3K-Akt. The 5-HT2A receptors expressed on glutamatergic cortical pyramidal cells and deep cortical layers (V and VI) are involved in the regulation of behavioral responses to numerous psychotropic agents [180,181]. Serotoninergic neurotransmission mediated by 5-HT2A receptors is a key target for therapy of sensory and cognitive disorders [182,183]. Interestingly, both 5-HT2A and 5-HT2B receptor antagonists [11,158] and 5-HT2CR [159] agonists have been used in attempts to correct AD-related conditions.

5.3. 5-HT3 Receptors

The 5-HT3 receptor is a ligand-gated ion channel. Homomeric and heteromeric 5-HT3 receptors mediate a rapidly activating, desensitizing, inward current, which is predominantly carried by sodium and potassium ions [184,185]. 5-HT3R is a homopentamer or heteropentamer and composed of five subunits, namely 5-HT3RA, 5-HT3RB, 5-HT3RC, 5-HT3RD, and 5- HT3RE [186,187]. The receptors are found both pre- and postsynaptically, and activation can modulate the release of a range of neurotransmitters, including dopamine, GABA, substance P, and acetylcholine [188,189,190].
Neurons in the neocortex, olfactory cortex, hippocampus, and amygdala that express the 5-HT3 receptor are primarily GABAergic neurons [191]. Approximately 50% of hippocampal interneurons express the 5-HT3 receptor [192,193]. In various animal models of AD, early Aβ plaques in the hippocampus are present exclusively in areas occupied by interneurons [194]. The 3A subunit of the 5-HT3 receptor was found to be significantly expressed in the brains of mice (AD model) and AD patients. In the mouse model, 5-HT3RA-positive interneurons were also clustered around Aβ plaques [195]. Treatment of mice with tropisetron, a 5-HT3R antagonist, for 8 weeks resulted in the partial reversal of cognitive deficits, and Aβ plaques and neuroinflammation were significantly reduced. These data led the authors to suggest that the inhibition of 5-HT3R GABAergic neurons at the initial stage of AD partially reverses the pathologic changes caused by the disease [195].

5.4. 5-HT4 Receptors

5-HT4R is an excitatory Gs-coupled receptor that activates cAMP synthesis, the cAMP-PKA pathway, and increases neuronal excitability. Ten different splice variants (5-HT4a, 5-HT4b, 5-HT4c, 5-HT4d, 5-HT4e, 5-HT4f, 5-HT4g, 5-HT4hb, 5-HT4i, and 5-HT4n) have been found in humans [196]. 5-HT4R is widely expressed throughout the CNS and plays an important role in the regulation of mood, anxiety, and cognitive function, and drugs that activate this receptor have rapid antidepressant effects [196,197]. Loss of functional 5-HT4R in excitatory hippocampal neurons leads to persistent AD-like behavioral responses and increased anxiety [198]. 5-HT4R is required to maintain proper excitability of granular cells of the dentate gyrus (DG) [199,200]. 5-HT4 agonists may be used to treat cognitive impairment associated with depression. The activation of 5-HT4 receptors may improve cognitive symptoms due to their known ability to enhance acetylcholine release and to some extent compensate for the deficit in cholinergic transmission in AD [201]. In preclinical studies, short-term treatment with 5-HT4R agonists (RS67333 and others) had anxiolytic and antidepressant properties and mimicked the cellular and molecular responses of AD induced by chronic SSRI administration [199,200]. The 5-HT4 receptor has also been shown to be a target for the treatment of MDD, and the pharmacologic stimulation of these receptors has been shown to improve learning and memory in healthy subjects [202].

5.5. 5-HT5 Receptors

The 5-HT5 subtypes are metabotropic receptors that activate the Gi/Go protein, suppress AC activity, and reduce the level of cAMP. The 5-HT5 subfamily consists of two members, designated as 5-HT5A and 5-HT5B. 5-HT5A receptors are expressed in the cerebral cortex, hippocampus, and raphe nuclei [203]. They exhibit antinociceptive properties and are involved in the regulation of memory and learning [204]. Attempts have also been undertaken to use them as targets in the treatment of Huntington’s disease and AD [162].

5.6. 5-HT6 Receptor

Among the known serotonin receptor subtypes, 5-HT6R is the only one whose expression is restricted to the CNS. The 5-HT6 subtypes are metabotropic receptors that are conjugated to a Gs protein that activates AC and increases cAMP levels. The 5-HT6 receptor is known to not only activate AC but also interacts with Fyn kinase, mediating the Ras-MEK-ERK1/2 signaling pathway, and with c-Jun activation domain binding protein-1 (Jab1), which binds to the c-Jun transcription factor [205,206,207]. During development, 5-HT6 signaling regulates the migration of cortical pyramidal neurons and interneurons and morphogenesis of the dendritic tree [208,209]. In vivo studies on the developing cortex have identified the involvement of 5-HT6 receptors in dendritic growth and differentiation of neurons that transmit signals via the Fyn pathway [210,211].
5-HT6 receptors are expressed in different brain regions [212,213]. Studies on the localization of 5-HT6R in the hippocampus have shown that they are mainly expressed on excitatory pyramidal cells and to a much lesser extent on inhibitory GABAergic interneurons [214,215]. The application of 5-HT6 receptor antagonists increases the levels of acetylcholine and glutamate in the frontal cortex and hippocampus resulting in enhanced excitatory neurotransmission. 5-HT6 receptor antagonists inhibit the mTOR complex, which promotes neuronal survival and increases neurite outgrowth [216]. This modulation of the 5-HT6 complex by mTOR suggests a potential for the treatment of anxiety, schizophrenia, and AD [163].
In AD, a significant decrease in 5-HT6R density was found in the cortical regions of patients [217]. Preclinical studies on rodents and primates have reported that 5-HT6R antagonists improve cognitive function in a wide variety of learning and memory paradigms and account for the effects of antidepressants [218,219]. The absence of 5-HT6R receptors in peripheral organs makes it a preferable therapeutic target for treating the cognitive symptoms of schizophrenia, autism spectrum disorders, and dementia associated with AD. The efficacy of 5-HT6R antagonists in alleviating cognitive impairment has been demonstrated in a number of rodent models of neurodegenerative, psychiatric, and neurodegenerative diseases [220,221,222,223].

5.7. 5-HT7 Receptors

Different isoforms of 5-HT7R are expressed in humans: 5-HT7R(A), 5-HT7R(B), and 5-HT7R(D) [224]. The activation of 5-HT7 receptors initiates signaling not only through Gαs [225] but also activates RhoA and Cdc42 (the non-canonical signaling pathway of 5-HT7R acts via Gα12) [226]. This leads to the activation of Rho, Rac, and cell division control protein 42 (Cdc42); all of them are part of the Rho family of small GTPases, which in neurons promote dendrite sprouting, the formation of filopodia, and synaptogenesis [227,228,229,230]. Moreover, the 5-HT7 receptor can regulate serum response element (SRE)-mediated gene transcription as well as modulate the morphofunctional state of neurons through the Gα12-dependent activation of RhoA and Cdc42 GTPases [231]. A critical role of 5-HT7R in neuronal morphogenesis has been demonstrated: serotonin activation of 5-HT7R leads to increased dendritic spike density and enhanced synaptogenesis in forebrain neurons [232]. 5-HT7R is widely expressed in the CNS, the gastrointestinal tract, and other organs, where it potentially regulates various physiological functions, including the sleep–wake cycle, learning and memory, body temperature, nociception [233,234,235], and depressive-like behavior [236,237]. In the CNS, 5-HT7R is expressed in different cell types, including neurons, astrocytes, and microglia [226,238,239]. LP-211, a highly selective 5-HT7R agonist, ameliorated neuronal damage and cognitive impairment induced by Aβ [164]. In a streptozotocin-mediated murine model of AD neurodegeneration, intracerebroventricular treatment with the AS-19, 5-HT7R selective agonist reduced LTP impairment and apoptosis in the hippocampus [240]. In this way, 5-HT7R agonists may be neuroprotective by acting at multiple levels, including the reduction in excitotoxicity and oxidative stress, synaptic remodeling, regulation of neurotrophic factors, or immunomodulation.

6. Serotonin Level, Inflammation, and Immunity

As mentioned in Table 1, various 5-HT receptors are presented on the cells of the cardiovascular system, namely on smooth muscle and endothelial cells, but they also are presented on the various, at least 11, types of immune cells [241], leading to the serotonin level, inflammation, and immunity being interconnected in complex ways. Serotonin acts as a signaling molecule in both the central nervous system and peripheral immune system. Serotonin regulates both pro- and anti-inflammatory immune responses. Peripheral 5-HT regulates immune cell activity (e.g., T cells, macrophages, and dendritic cells).
Serotonin action on immune cells is multifaceted. 5-HT can enhance inflammation by activating Th1/Th17 cells and promoting the release of cytokines (e.g., TNF-α, IL-6) [242,243]. At other times, 5-HT may suppress inflammation by increasing the activity of regulatory T cells [244]. Gut-produced 5-HT influences systemic inflammation, which can affect brain depression [245]. Chronic inflammation from obesity or stress lowers 5-HT, which exacerbates mood disorders [246]. Reduced 5-HT levels have been found also in autoimmune diseases (e.g., rheumatoid arthritis, IBD) [247]. Pro-inflammatory cytokines (e.g., IL-1β, IL-6) can reduce serotonin levels [248] by reducing 5-HT precursor tryptophan via the kynurenine pathway (converting it to neurotoxic metabolites) [249]. Treatments targeting inflammation (e.g., anti-cytokine drugs) may help serotonin-related disorders. Chronic stress suppression reduces serum cortisol levels and inflammation, and increases 5-HT levels [250].
It should be noted that the inflammatory process in the CNS and the pathogenesis of AD are interrelated: the intensity of neuroinflammation increases as a result of neurodegeneration and Aβ deposition, which contributes to the strengthening of the same processes. As one of the mechanisms, it has been shown that an increase in the concentration of proinflammatory cytokines depresses melatonin synthesis in the pineal gland, which leads to the insufficiency of its effects, in particular, to the impaired regulation of circadian rhythms [251]. Furthermore, sleep deprivation leads to an increased production of proinflammatory cytokines, such as IL-1β, IL-6, and IL-17 [252]. As a result, a vicious circle between circadian rhythm regulation and neuroinflammation is formed.

7. Conclusions: Serotonin Is a Factor of Healthy Longevity

This review focuses on the involvement of the 5-HT system in the pathological processes leading to the development of AD. Figure 3 shows that the dysfunction of the 5-HT system causes AD progression through the disruption of circadian rhythms and the glymphatic system, Aβ plaque formation, mitochondrial dysfunction, neurodegeneration, decreased neurogenesis, depression, and stress. New data on the role of circadian rhythm disruption in glymphatic system dysfunction and Aβ accumulation in the brain, modulation of stress, depression, and the 5-HT system itself are discussed.
The 5-HT system remains a central therapeutic target in AD. Preclinical and clinical studies suggest that serotonergic dysfunction contributes to Aβ accumulation, neuroinflammation, tau pathology, and cognitive decline. Early loss of serotonergic neurons in the raphe nuclei precedes cognitive symptoms. The 5-HT system’s complexity and central role in multiple disorders make it a prime target for novel therapeutics, especially in mental health and neurology. Selective serotonin reuptake inhibitors (SSRIs) (e.g., citalopram, sertraline) remain first-line therapy but require dose adjustments due to altered pharmacokinetics in aging. Chronic SSRI use leads to the desensitization of these receptors, enhancing 5-HT release. SSRIs show slow Aβ accumulation in the brain [253] and decrease depression and anxiety. Anti-inflammatory agents (e.g., non-steroidal anti-inflammatory drugs and statins) may synergize with SSRIs.
Aging is associated with decreased 5-HT synthesis, reduced receptor density (e.g., 5-HT1A and 5-HT2A), and impaired synaptic plasticity, all of which contribute to late-life depression (LLD). LLD increases the risk of AD via amyloid-β accumulation and the loss of 5-HT neurons in the raphe nuclei [12]. The activation of postsynaptic 5-HT1A receptors promotes neurogenesis and resilience to stress, which are critical for LLD with cognitive impairment. 5-HT4 receptor agonists show promise in reducing amyloid pathology as well. Exercise and a Mediterranean diet increase 5-HT and BDNF levels, thereby improving mood and cognition. However, the role of the 5-HT system in LLD extends beyond monoamine deficiency and involves receptor dynamics, neuroinflammation, and vascular pathology, requiring tailored approaches.
The etiology and pathogenesis of neurodegenerative diseases, including AD, are multifactorial; therefore, one treatment strategy is to develop drugs that can act on multiple targets involved in disease pathogenesis. Increasing evidence suggests that enhancing the global antioxidant defense system may be more effective in controlling oxidative stress-related pathogenesis in the AD brain [254].
Serotonin levels are closely related to inflammation. On the one hand, 5-HT can suppress systemic inflammation, which affects brain depression [242,248]. On the other hand, chronic inflammation caused by obesity or stress reduces 5-HT levels, which exacerbates psychiatric disorders. Thus, treatments aimed at suppressing inflammation (e.g., anti-cytokine drugs) may help with 5-HT deficiency disorders. Anti-inflammatory diets (omega-3 fatty acids, polyphenols) can also improve 5-HT production and maintain 5-HT levels.
Hybrid polyfunctional molecules combining anticholinesterase activity and a high affinity for G-protein-coupled receptors appear promising for the treatment of AD. Thus, a series of such polyfunctional agents have been synthesized and tested: cholinesterase inhibitors possessing high selectivity to the type 3 histamine receptor [255], on cannabinoid type 2receptors [256,257] as well as on serotonin receptors: 5-HT6 [258] and 5-HT4 [259].
One of the first 5-HT4 receptor agonists of the benzimidazolone class is BIMU-8, which is able to cross the blood–brain barrier. It attenuates respiratory depression by activating the pre-Bötzinger complex located in the respiratory center of the brainstem [260]. The compound has also been shown to increase brain activity and improve learning and memory [261].
Multifunctional ligands are intended for use in both symptomatic and pathogenetic therapy. Nevertheless, the utilization of this approach is accompanied by a quantity of interrelated problems: (a) high molecular weight of “hybrid” molecules; (b) inability to cross the blood–brain barrier; (c) low bioavailability during oral administration; (d) the need to optimize selectivity and affinity for different targets; and (e) action of “hybrid” molecule components at different concentration ranges. It is essential to take into account these peculiarities at the very first stages to make appropriate computational studies before proceeding to the animal research. Recently, an algorithm has been proposed to design “functional binaries” that combine the properties of cholinesterase inhibitors and 5-HTR activators. At the first stage, a group of substituted 1,3-dihydro-2-oxy-1H-benzimidazol-2-ones was formed for further synthesis. Using Percepta software, logP, logBB, and LD50 (mg/kg) were theoretically calculated for mice and rats. Based on the data obtained, the most promising compounds were selected, which were subsequently synthesized and their anticholinesterase activity was investigated under in vitro conditions [262]. The biochemical analysis performed showed the ability of the obtained structures to inhibit AChE and BChE. In the next stage of the experimental research, the interaction of several substituted 1,3-dihydro-2-oxo-1H-benzimidazol-2-ones with three types of Gs-protein-coupled serotonin receptors, 5-HT6, 5-HT4, and 5-HT7, was investigated in silico. Molecular modeling methods, such as molecular docking and molecular dynamics simulations in water and a lipid bilayer, were used to study the interaction of the compounds with 5-HTR. In addition, molecular modeling methods were used to investigate the mechanism of interaction of the tested compounds with cholinesterases to describe the binding sites, and to reveal the structural features of the drugs that determine the potency of their anticholinesterase activity. A primary in silico evaluation showed that benzimidazole–carboxamides effectively bind to 5-HT4R and 5-HT7R. The pool of the obtained data allows us to choose N-[2-(diethylamino)ethyl]-2-oxo-3-(tert-butyl)-2,3-dihydro-1H-benzimidazole-1-carboxamide hydrochloride as the most promising for further experimental development [263].
Physical exercise is known to have positive effects on brain function. For example, walking, especially high-intensity walking, is associated with improved episodic memory, even when started in middle age [264]. Exercise improves mood by enhancing mitochondrial function and neuroplasticity in the dorsal raphe [265]. Multicomponent training programs may be an important non-pharmacologic strategy to improve physical and cognitive function in hospitalized AD patients [266]. Short-term aggravated exercise reduced neuroinflammation and attenuated neuropathological changes in AD mice [267]. Combined multimodal exercise is superior to aerobic and weight-bearing exercise in terms of stimulating executive function [268]. Regular aerobic exercise can increase serotonin release and modulate synaptic plasticity in the anterior cingulate cortex, ultimately reducing pain and associated anxiety behaviors through the functions of serotonin 5-HT1A and 5-HT7 receptors [267].
Increasing evidence indicates a strong association between a poor diet and the exacerbation of mood disorders, including anxiety and depression, as well as other neuropsychiatric conditions. The current epidemiological data on nutrition and mental health do not provide information about causality or underlying mechanisms; therefore, an experimental medicine approach and a mechanistic understanding are required to provide solid evidence on which future policies on diet and nutrition for mental health can be based [269]. As no drugs are available to prevent the progression of these neurological disorders, intervention strategies using phytochemicals have been proposed as an alternative form of treatment. Some researchers consider organosulfur compounds as ideal nutraceutical agents as they can serve not only as direct antioxidants trapping electrons, but also have non-antioxidant effects, such as antiplatelet, fibrinolytic, anti-inflammatory, immunomodulatory, anti-aging actions, etc. [270]. Among phytochemicals, isothiocyanate sulforaphane has demonstrated neuroprotective effects in several in vitro and in vivo studies. In particular, the evidence suggests that sulforaphane’s beneficial effects can be mainly ascribed to its peculiar ability to activate the Nrf2/ARE pathway [271]. Sulforaphane has also been shown to stimulate ubiquitin proteasome system (UPS) activity in vitro; moreover, it activates protein degradation machineries (autophagic activities) in both the brain and peripheral tissues [271]. S-allylcysteine (SAC) prevents the progression of AD, and there are multiple mechanisms underlying this ability [272]. Given that Nrf2 is a major switch in the expression of most antioxidant enzymes and PGC-1α, the activation of Nrf2 appears to be a good therapeutic strategy to control oxidative stress in the brain of AD patients. FGF, flavonoids, α-lipoic acid, allicin, and taurine appear to be able to activate the Nrf2-ARE antioxidant defense system [273,274,275,276].
Accumulating data indicate that sulfane sulfur has important functions in cells. The broad diversity of effects suggests that its functions are general and not specific to any tissue or any process. Moreover, it should not be called a “signaling agent” since there is no evidence that it acts in a controlled rise and fall pattern (as with neurotransmitters or hormones) [277]. Both redox sensing and redox signaling use sulfur switches, especially Cys residues in proteins, which are sensitive to reversible oxidation, nitrosylation, glutathionylation, acylation, sulfhydration, or metal binding. Attempts to regulate redox state have focused on electrophiles, which activate potent cellular defense systems against oxidative stress. Nutraceuticals can serve not only as electrophiles, but as a kind of “pro-electrophilic drug” that becomes electrophilic in response to oxidation and then activates the Keap1/Nrf2/ARE transcription pathway to synthesize endogenous antioxidant “phase 2” enzymes [278]. Another important prerequisite of the new concept in redox biology/medicine is the use of compounds that has been termed “pathologically activated therapeutics”, i.e., relatively small molecules are chemically converted to their active form by the very oxidative stress that they are intended to then combat [278].
Summarizing our multifaceted analysis of the role of serotonergic regulation in the pathogenesis of development and treatment options for AD, we come to the generally banal conclusion that the combination of physical activity, positive emotions, and multimodal medications that modulate the serotonergic system is the key to healthy longevity.

Author Contributions

Conceptualization, L.P.D., V.P.Z. and N.V.G.; writing—original draft preparation, L.P.D., V.P.Z. and N.V.G.; writing—review and editing, V.P.Z., A.D.N. and N.V.G.; supervision, N.V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the State Reserves No. 075-00263-25-00 of the Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences (to N.V.G.), and by the State Reserves No. FMRM-2025-0027 of the Ministry of Science and Higher Education of the Russian Federation (to L.P.D., V.P.Z., A.D.N.). The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
5HTSerotonin
5-HT systemSerotonergic system
ADAlzheimer’s disease
Amyloid β
AβOAmyloid β oligomers
ACAdenylate cyclase
ACCAnterior cingulate cortex
AChEAcetylcholinesterase
APPAmyloid precursor protein
sAPPαSecreted form of APP
AQP4Aquaporin-4
BChEButyrylcholinesterase
BDNFBrain-derived neurotrophic factor
CNSCentral nervous system
CVSCardiovascular system
DRNDorsal raphe nucleus
ETCElectron transport chain
FGFFibroblast growth factor
GABAγ-aminobutyric acid
LTDLong-term depression
LTPLong-term potentiation
mAHPMedium-duration afterhyperpolarization
MDDMajor depressive disorder
mPTPMitochondrial permeability transition pore
MRNMedian raphe nucleus
NMDARNMDA receptor
Nrf2Nuclear respiratory factor 2
PANoptosisPyroptosis, apoptosis, and necroptosis
PFCPrefrontal cortex
PNSPeripheral nervous system
PrPcCellular prion protein
pTauTau protein
ROSReactive oxygen species
SCNSuprachiasmatic nucleus
SERTSerotonin transporter
SSRIsSelective serotonin reuptake inhibitors
TIMTranslocase of the inner membrane
TOMTranslocase of the outer membrane

Appendix A. Chemical Names for the Research Compounds

  • 5-HTP-DP—N-acetyl-5-hydroxytrytophyl-5-hydroxytryptophan amide.
  • 8-OH-DPAT—7-(Dipropylamino)-5,6,7,8-tetrahydronaphthalen-1-ol.
  • AS-19—(2S)-5-(1,3,5-Trimethylpyrazol-4-yl)-2-(dimethylamino)tetralin.
  • BIMU1—33-ethyl-N-[(5S)-8-methyl-8-azabicyclo [3.2.1]octan-3-yl]-2-oxobenzimidazole-1-carboxamide.
  • BIMU8—N-[(1R,5S)-8-methyl-8-azabicyclo [3.2.1]oct-3-yl]-2-oxo-3-(propan-2-yl)-2,3-dihydro-1H-benzimidazole-1-carboxamide hydrochloride.
  • BP 554—1-[3-(3,4-methylenedioxyphenoxy)propyl]-4-phenyl-piperazine maleate.
  • BRL15572—3-(4-(3-chlorophenyl)piperazin-1-yl)-1,1-diphenyl-2-propanol.
  • BRL54443—5-Hydroxy-3-(1-methylpiperidin-4-yl)-1H-indole.
  • BW723C86—α-Methyl-5-(2-thienylmethoxy)-1H-indole-3-ethanamine hydrochloride.
  • CGS 12066B—7-Trifluoromethyl-4-(4-methyl-1-piperazinyl)pyrrolo [1,2-a]-quinoxalinedimaleate.
  • CP-93129—1,4-Dihydro-3-(1,2,3,6-tetrahydro-4-pyridinyl)-5H-pyrrol [3,2-b]pyridin-5-one dihydrochloride.
  • DOI—2,5-Dimethoxy-4-iodoamphetamine.
  • EMD386088—5-Chloro-2-methyl-3-(1,2,3,6-tetrahydro-4-pyridinyl)-1H-indole.
  • GR113808—[1-[2-[(Methylsulfonyl)amino]ethyl]-4-piperidinyl]methyl 1-methyl-1H-indole-3-carboxylate.
  • GR-127935—N-[4-methoxy-3-(4-methyl-1-piperazinyl)phenyl]-2′-methyl-4′-(5-methyl-1,2,4-oxadiazol-3-yl)-1-1′-biphenyl-4-carboxamide.
  • GR 4661—3-[3-(2-Dimethylaminoethyl)-1H-indol-5-yl]-N-(4-methoxybenzyl)acrylamid.
  • GR55562—3-[3-(Dimethylamino)propyl]-4-hydroxy-N-[4-(4-pyridinyl)phenyl]benzamide dihydrochloride.
  • L-703,664—2-((3-(2-(dimethylamino)ethyl)-1H-indol-5-yl)methyl)-5-methyl-1,2,5-thiadiazolidine 1,1-dioxide succinate.
  • LP12—4-(1,1′-Biphenyl)-2-yl-N-(1,2,3,4-tetrahydro-1-naphthalenyl)-1-piperazinehexanamide hydrochloride.
  • LP44—6-[4-(2-methylsulfanylphenyl)piperazin-1-yl]-N-(1,2,3,4-tetrahydronaphthalen-1-yl)hexanamide hydrochloride.
  • LP211—4-[1,1′-Biphenyl]-2-yl-N-[(4-cyanophenyl)methyl]-1-piperazinehexanamide.
  • LY272015—1-[(3,4-dimethoxyphenyl)methyl]-6-methyl-2,3,4,9-tetrahydro-1H-pyrido [3,4-b]indole hydrochloride.
  • LY310762—1-[2-[4-(4-fluorobenzoyl)piperidin-1-yl]ethyl]-3,3-dimethylindol-2-one hydrochloride.
  • LY334370—4-fluoro-N-[3-(1-methylpiperidin-4-yl)-1H-indol-5-yl]benzamide.
  • LY344864—N-[(6R)-6-(dimethylamino)-6,7,8,9-tetrahydro-5H-carbazol-3-yl]-4-fluorobenzamide.
  • MDL72222—3-tropanyl-3,5-dichlorobenzoat.
  • MK212—2-chloro-6-piperazin-1-ylpyrazine hydrochloride.
  • NAN190—1-(2-Methoxyphenyl)-4-(4-phthalimidobutyl)piperazine hydrobromide.
  • NAS-181—(2R)-2-[[[3-(4-morpholinylmethyl)-2H-1-benzopyran-8-yl]oxy]methyl]-morpholine bimethanesulfonate.
  • PNU-109291—(S)-3,4-Dihydro-1-[2-[4-(4-methoxyphenyl)-1-piperazinyl]ethyl]-N-methyl-1H-2-benzopyran-6-carboxamide.
  • R-96544—(2R,4R)-5-[2-[2-[2-(3-Methoxyphenyl)ethyl]phenoxy]ethyl]-1-methyl-3-pyrrolidinol hydrochloride.
  • Ro 04-6790—4-amino-N-[2,6-bis(methylamino)pyrimidin-4-yl]benzenesulfonamide.
  • RS100235—1-(5-amino-6-chloro-2,3-dihydro-1,4-benzodioxin-8-yl)-3-[1-[3-(3,4-dimethoxyphenyl)propyl]piperidin-4-yl]propan-1-one.
  • RS102221—N-[5-[5-(2,4-dioxo-1,3,8-triazaspiro [4.5]decan-8-yl)pentanoyl]-2,4-dimethoxyphenyl]-4-(trifluoromethyl)benzenesulfonamide.
  • RS127445—4-(4-fluoronaphthalen-1-yl)-6-propan-2-ylpyrimidin-2-amine.
  • RS67333—1-(4-amino-5-chloro-2-methoxyphenyl)-3-(1-butylpiperidin-4-yl)propan-1-one.
  • RS67506—N-[2-[4-[3-(4-amino-5-chloro-2-methoxyphenyl)-3-oxopropyl]piperidin-1-yl]ethyl]methanesulfonamide hydrochloride.
  • SB204070—(1-butylpiperidin-4-yl)methyl 5-amino-6-chloro-2,3-dihydro-1,4-benzodioxine-8-carboxylate.
  • SB236057—(1′-ethylspiro [6,7-dihydro-2H-furo [2,3-f]indole-3,4′-piperidine]-5-yl)-[4-[2-methyl-4-(5-methyl-1,3,4-oxadiazol-2-yl)phenyl]phenyl]methanone.
  • SB242084—6-chloro-5-methyl-N-[6-(2-methylpyridin-3-yl)oxypyridin-3-yl]-2,3-dihydroindole-1-carboxamide.
  • SB258719—(1R)-3,N-dimethyl-N-[1-methyl-3-(4-methylpiperidin-1-yl)propyl]benzenesulfonamide.
  • SB269970—3-[(2R)-2-[2-(4-methylpiperidin-1-yl)ethyl]pyrrolidin-1-yl]sulfonylphenol.
  • SB271046—5-chloro-N-(4-methoxy-3-piperazin-1-ylphenyl)-3-methyl-1-benzothiophene-2-sulfonamide.
  • SB272183—5-chloro-6-(4-methylpiperazin-1-yl)-N-(4-pyridin-4-ylnaphthalen-1-yl)-2,3-dihydroindole-1-carboxamide.
  • SB399885—N-(3,5-dichloro-2-methoxyphenyl)-4-methoxy-3-piperazin-1-ylbenzenesulfonamide.
  • SB699551—3-cyclopentyl-N-[2-(dimethylamino)ethyl]-N-[[4-[4-[(2-phenylethylamino)methyl]phenyl]phenyl]methyl]propanamide;dihydrochloride.
  • SR57227A—1-(6-chloropyridin-2-yl)piperidin-4-amine.
  • U92016A—(8R)-8-(dipropylamino)-6,7,8,9-tetrahydro-3H-benzo[e]indole-2-carbonitrile
  • WAY 100635—N-[2-[4-(2-Methoxyphenyl)-1-piperazinyl]ethyl]-N-(2-pyridyl)cyclohexanecarboxamide.
  • WAY 163909—(11R,15R)-7,10-diazatetracyclo [8.5.1.05,16.011,15]hexadeca-1,3,5(16)-triene.
  • WAY 181187—2-[1-(6-chloroimidazo [2,1-b][1,3]thiazol-5-yl)sulfonylindol-3-yl]ethanamine.

References

  1. Rodríguez, J.J.; Noristani, H.N.; Verkhratsky, A. The Serotonergic System in Ageing and Alzheimer’s Disease. Prog. Neurobiol. 2012, 99, 15–41. [Google Scholar] [CrossRef] [PubMed]
  2. Eremin, D.V.; Kondaurova, E.M.; Rodnyy, A.Y.; Molobekova, C.A.; Kudlay, D.A.; Naumenko, V.S. Serotonin Receptors as a Potential Target in the Treatment of Alzheimer’s Disease. Biochem. Biokhimiia 2023, 88, 2023–2042. [Google Scholar] [CrossRef] [PubMed]
  3. Butzlaff, M.; Ponimaskin, E. The Role of Serotonin Receptors in Alzheimer’s Disease. Opera Medica Physiol. 2016, 2, 91–100. [Google Scholar] [CrossRef]
  4. Päivärinta, M.A.; Marttila, R.J.; Lönnberg, P.; Rinne, U.K. Decreased Raphe Serotonin in Rabbits with Experimental Herpes Simplex Encephalitis. Neurosci. Lett. 1993, 156, 1–4. [Google Scholar] [CrossRef]
  5. Melnikov, M.; Sviridova, A.; Rogovskii, V.; Oleskin, A.; Boziki, M.; Bakirtzis, C.; Kesidou, E.; Grigoriadis, N.; Boyko, A. Serotoninergic System Targeting in Multiple Sclerosis: The Prospective for Pathogenetic Therapy. Mult. Scler. Relat. Disord. 2021, 51, 102888. [Google Scholar] [CrossRef]
  6. Carneiro, I.B.C.; Toscano, A.E.; Da Cunha, M.D.S.B.; Lacerda, D.C.; Pontes, P.B.; De Castro, R.M.; De Jesus Deiró, T.C.B.; Medeiros, J.M.B. Serotonergic Mechanisms Associated with Experimental Models of Hypoxia: A Systematic Review. Int. J. Dev. Neurosci. 2022, 82, 667–679. [Google Scholar] [CrossRef]
  7. Tian, J.; Stucky, C.S.; Wang, T.; Muma, N.A.; Johnson, M.; Du, H. Mitochondrial Dysfunction Links to Impaired Hippocampal Serotonin Release in a Mouse Model of Alzheimer’s Disease. J. Alzheimers Dis. JAD 2023, 93, 605–619. [Google Scholar] [CrossRef]
  8. Cardon, I.; Grobecker, S.; Jenne, F.; Jahner, T.; Rupprecht, R.; Milenkovic, V.M.; Wetzel, C.H. Serotonin Effects on Human iPSC-Derived Neural Cell Functions: From Mitochondria to Depression. Mol. Psychiatry 2024, 29, 2689–2700. [Google Scholar] [CrossRef]
  9. Selkoe, D.J.; Hardy, J. The Amyloid Hypothesis of Alzheimer’s Disease at 25 Years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef]
  10. Burstein, E.S. Relevance of 5-HT2A Receptor Modulation of Pyramidal Cell Excitability for Dementia-Related Psychosis: Implications for Pharmacotherapy. CNS Drugs 2021, 35, 727–741. [Google Scholar] [CrossRef]
  11. Afshar, S.; Shahidi, S.; Rohani, A.H.; Soleimani Asl, S.; Komaki, A. Protective Effects of 5-HT1A Receptor Antagonist and 5-HT2A Receptor Agonist on the Biochemical and Histological Features in a Rat Model of Alzheimer’s Disease. J. Chem. Neuroanat. 2019, 96, 140–147. [Google Scholar] [CrossRef]
  12. Bernedo, V.; Insua, D.; Suárez, M.-L.; Santamarina, G.; Sarasa, M.; Pesini, P. Beta-Amyloid Cortical Deposits Are Accompanied by the Loss of Serotonergic Neurons in the Dog. J. Comp. Neurol. 2009, 513, 417–429. [Google Scholar] [CrossRef]
  13. Song, N.-N.; Huang, Y.; Yu, X.; Lang, B.; Ding, Y.-Q.; Zhang, L. Divergent Roles of Central Serotonin in Adult Hippocampal Neurogenesis. Front. Cell. Neurosci. 2017, 11, 185. [Google Scholar] [CrossRef] [PubMed]
  14. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Chaimani, A.; Atkinson, L.Z.; Ogawa, Y.; Leucht, S.; Ruhe, H.G.; Turner, E.H.; Higgins, J.P.T.; et al. Comparative Efficacy and Acceptability of 21 Antidepressant Drugs for the Acute Treatment of Adults with Major Depressive Disorder: A Systematic Review and Network Meta-Analysis. Lancet Lond. Engl. 2018, 391, 1357–1366. [Google Scholar] [CrossRef]
  15. Funk, K.A.; Bostwick, J.R. A Comparison of the Risk of QT Prolongation among SSRIs. Ann. Pharmacother. 2013, 47, 1330–1341. [Google Scholar] [CrossRef]
  16. Noristani, H.N.; Verkhratsky, A.; Rodríguez, J.J. High Tryptophan Diet Reduces CA1 Intraneuronal β-Amyloid in the Triple Transgenic Mouse Model of Alzheimer’s Disease. Aging Cell 2012, 11, 810–822. [Google Scholar] [CrossRef] [PubMed]
  17. Citron, M. Alzheimer’s Disease: Strategies for Disease Modification. Nat. Rev. Drug Discov. 2010, 9, 387–398. [Google Scholar] [CrossRef]
  18. Olajide, O.J.; Suvanto, M.E.; Chapman, C.A. Molecular Mechanisms of Neurodegeneration in the Entorhinal Cortex That Underlie Its Selective Vulnerability during the Pathogenesis of Alzheimer’s Disease. Biol. Open 2021, 10, bio056796. [Google Scholar] [CrossRef] [PubMed]
  19. Feng, W.; Zhang, Y.; Wang, Z.; Xu, H.; Wu, T.; Marshall, C.; Gao, J.; Xiao, M. Microglia Prevent Beta-Amyloid Plaque Formation in the Early Stage of an Alzheimer’s Disease Mouse Model with Suppression of Glymphatic Clearance. Alzheimers Res. Ther. 2020, 12, 125. [Google Scholar] [CrossRef]
  20. d’Errico, P.; Meyer-Luehmann, M. Mechanisms of Pathogenic Tau and Aβ Protein Spreading in Alzheimer’s Disease. Front. Aging Neurosci. 2020, 12, 265. [Google Scholar] [CrossRef]
  21. Zhang, H.; Wei, W.; Zhao, M.; Ma, L.; Jiang, X.; Pei, H.; Cao, Y.; Li, H. Interaction between Aβ and Tau in the Pathogenesis of Alzheimer’s Disease. Int. J. Biol. Sci. 2021, 17, 2181–2192. [Google Scholar] [CrossRef] [PubMed]
  22. Atlante, A.; Valenti, D.; Latina, V.; Amadoro, G. Role of Oxygen Radicals in Alzheimer’s Disease: Focus on Tau Protein. Oxygen 2021, 1, 96–120. [Google Scholar] [CrossRef]
  23. Tönnies, E.; Trushina, E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. J. Alzheimers Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef]
  24. Rhein, V.; Song, X.; Wiesner, A.; Ittner, L.M.; Baysang, G.; Meier, F.; Ozmen, L.; Bluethmann, H.; Dröse, S.; Brandt, U.; et al. Amyloid-Beta and Tau Synergistically Impair the Oxidative Phosphorylation System in Triple Transgenic Alzheimer’s Disease Mice. Proc. Natl. Acad. Sci. USA 2009, 106, 20057–20062. [Google Scholar] [CrossRef]
  25. Spuch, C.; Ortolano, S.; Navarro, C. New Insights in the Amyloid-Beta Interaction with Mitochondria. J. Aging Res. 2012, 2012, 324968. [Google Scholar] [CrossRef]
  26. Reddy, P.H.; Oliver, D.M. Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer’s Disease. Cells 2019, 8, 488. [Google Scholar] [CrossRef] [PubMed]
  27. Hu, Y.; Li, X.-C.; Wang, Z.; Luo, Y.; Zhang, X.; Liu, X.-P.; Feng, Q.; Wang, Q.; Yue, Z.; Chen, Z.; et al. Tau Accumulation Impairs Mitophagy via Increasing Mitochondrial Membrane Potential and Reducing Mitochondrial Parkin. Oncotarget 2016, 7, 17356–17368. [Google Scholar] [CrossRef] [PubMed]
  28. Du, H.; Yan, S.S. Mitochondrial Permeability Transition Pore in Alzheimer’s Disease: Cyclophilin D and Amyloid Beta. Biochim. Biophys. Acta 2010, 1802, 198–204. [Google Scholar] [CrossRef]
  29. Thinakaran, G.; Koo, E.H. Amyloid Precursor Protein Trafficking, Processing, and Function. J. Biol. Chem. 2008, 283, 29615–29619. [Google Scholar] [CrossRef]
  30. Haass, C.; Willem, M. Secreted APP Modulates Synaptic Activity: A Novel Target for Therapeutic Intervention? Neuron 2019, 101, 557–559. [Google Scholar] [CrossRef]
  31. Wilkins, H.M.; Swerdlow, R.H. Amyloid Precursor Protein Processing and Bioenergetics. Brain Res. Bull. 2017, 133, 71–79. [Google Scholar] [CrossRef] [PubMed]
  32. Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al. The Amyloid-β Pathway in Alzheimer’s Disease. Mol. Psychiatry 2021, 26, 5481–5503. [Google Scholar] [CrossRef] [PubMed]
  33. Gakhar-Koppole, N.; Hundeshagen, P.; Mandl, C.; Weyer, S.W.; Allinquant, B.; Müller, U.; Ciccolini, F. Activity Requires Soluble Amyloid Precursor Protein Alpha to Promote Neurite Outgrowth in Neural Stem Cell-Derived Neurons via Activation of the MAPK Pathway. Eur. J. Neurosci. 2008, 28, 871–882. [Google Scholar] [CrossRef] [PubMed]
  34. Tang, Y.; Wang, Y.; Gao, Z.; Li, J.; Zhang, L.; Shi, H.; Dong, J.; Song, S.; Qian, C. sAPPα Peptide Promotes Damaged Microglia to Clear Alzheimer’s Amyloid-β via Restoring Mitochondrial Function. Chem. Weinh. Bergstr. Ger. 2024, 30, e202400870. [Google Scholar] [CrossRef]
  35. Demars, M.P.; Hollands, C.; Zhao, K.D.T.; Lazarov, O. Soluble Amyloid Precursor Protein-α Rescues Age-Linked Decline in Neural Progenitor Cell Proliferation. Neurobiol. Aging 2013, 34, 2431–2440. [Google Scholar] [CrossRef]
  36. Shankar, G.M.; Li, S.; Mehta, T.H.; Garcia-Munoz, A.; Shepardson, N.E.; Smith, I.; Brett, F.M.; Farrell, M.A.; Rowan, M.J.; Lemere, C.A.; et al. Amyloid-β Protein Dimers Isolated Directly from Alzheimer’s Brains Impair Synaptic Plasticity and Memory. Nat. Med. 2008, 14, 837–842. [Google Scholar] [CrossRef]
  37. Townsend, M.; Shankar, G.M.; Mehta, T.; Walsh, D.M.; Selkoe, D.J. Effects of Secreted Oligomers of Amyloid Β-protein on Hippocampal Synaptic Plasticity: A Potent Role for Trimers. J. Physiol. 2006, 572, 477–492. [Google Scholar] [CrossRef]
  38. Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s Disease: The Challenge of the Second Century. Sci. Transl. Med. 2011, 3, 77sr1. [Google Scholar] [CrossRef]
  39. Pacheco-Quinto, J.; Clausen, D.; Pérez-González, R.; Peng, H.; Meszaros, A.; Eckman, C.B.; Levy, E.; Eckman, E.A. Intracellular Metalloprotease Activity Controls Intraneuronal Aβ Aggregation and Limits Secretion of Aβ via Exosomes. FASEB J. 2019, 33, 3758–3771. [Google Scholar] [CrossRef]
  40. Brewer, G.J.; Herrera, R.A.; Philipp, S.; Sosna, J.; Reyes-Ruiz, J.M.; Glabe, C.G. Age-Related Intraneuronal Aggregation of Amyloid-β in Endosomes, Mitochondria, Autophagosomes, and Lysosomes. J. Alzheimers Dis. 2020, 73, 229–246. [Google Scholar] [CrossRef]
  41. Hayden, E.Y.; Teplow, D.B. Amyloid β-Protein Oligomers and Alzheimer’s Disease. Alzheimers Res. Ther. 2013, 5, 60. [Google Scholar] [CrossRef]
  42. Bode, D.C.; Freeley, M.; Nield, J.; Palma, M.; Viles, J.H. Amyloid-β Oligomers Have a Profound Detergent-like Effect on Lipid Membrane Bilayers, Imaged by Atomic Force and Electron Microscopy. J. Biol. Chem. 2019, 294, 7566–7572. [Google Scholar] [CrossRef] [PubMed]
  43. Yasumoto, T.; Takamura, Y.; Tsuji, M.; Watanabe-Nakayama, T.; Imamura, K.; Inoue, H.; Nakamura, S.; Inoue, T.; Kimura, A.; Yano, S.; et al. High Molecular Weight Amyloid β 1-42 Oligomers Induce Neurotoxicity via Plasma Membrane Damage. FASEB J. 2019, 33, 9220–9234. [Google Scholar] [CrossRef]
  44. Kang, J.-E.; Lim, M.M.; Bateman, R.J.; Lee, J.J.; Smyth, L.P.; Cirrito, J.R.; Fujiki, N.; Nishino, S.; Holtzman, D.M. Amyloid-Beta Dynamics Are Regulated by Orexin and the Sleep-Wake Cycle. Science 2009, 326, 1005–1007. [Google Scholar] [CrossRef] [PubMed]
  45. Boespflug, E.L.; Iliff, J.J. The Emerging Relationship Between Interstitial Fluid-Cerebrospinal Fluid Exchange, Amyloid-β, and Sleep. Biol. Psychiatry 2018, 83, 328–336. [Google Scholar] [CrossRef]
  46. Quentin, E.; Belmer, A.; Maroteaux, L. Somato-Dendritic Regulation of Raphe Serotonin Neurons; A Key to Antidepressant Action. Front. Neurosci. 2018, 12, 982. [Google Scholar] [CrossRef] [PubMed]
  47. Green, R.C.; Cupples, L.A.; Kurz, A.; Auerbach, S.; Go, R.; Sadovnick, D.; Duara, R.; Kukull, W.A.; Chui, H.; Edeki, T.; et al. Depression as a Risk Factor for Alzheimer Disease: The MIRAGE Study. Arch. Neurol. 2003, 60, 753–759. [Google Scholar] [CrossRef]
  48. Swartz, J.R.; Miller, B.L.; Lesser, I.M.; Booth, R.; Darby, A.; Wohl, M.; Benson, D.F. Behavioral Phenomenology in Alzheimer’s Disease, Frontotemporal Dementia, and Late-Life Depression: A Retrospective Analysis. J. Geriatr. Psychiatry Neurol. 1997, 10, 67–74. [Google Scholar] [CrossRef]
  49. Mitolo, M.; Tonon, C.; La Morgia, C.; Testa, C.; Carelli, V.; Lodi, R. Effects of Light Treatment on Sleep, Cognition, Mood, and Behavior in Alzheimer’s Disease: A Systematic Review. Dement. Geriatr. Cogn. Disord. 2018, 46, 371–384. [Google Scholar] [CrossRef]
  50. Padovani, A.; Antonini, A.; Barone, P.; Bellelli, G.; Fagiolini, A.; Ferini Strambi, L.; Sorbi, S.; Stocchi, F. Exploring Depression in Alzheimer’s Disease: An Italian Delphi Consensus on Phenomenology, Diagnosis, and Management. Neurol. Sci. Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2023, 44, 4323–4332. [Google Scholar] [CrossRef]
  51. Rapp, M.A.; Schnaider-Beeri, M.; Purohit, D.P.; Perl, D.P.; Haroutunian, V.; Sano, M. Increased Neurofibrillary Tangles in Patients with Alzheimer Disease with Comorbid Depression. Am. J. Geriatr. Psychiatry Off. J. Am. Assoc. Geriatr. Psychiatry 2008, 16, 168–174. [Google Scholar] [CrossRef] [PubMed]
  52. Hirschfeld, R.M. History and Evolution of the Monoamine Hypothesis of Depression. J. Clin. Psychiatry 2000, 61 (Suppl. 6), 4–6. [Google Scholar] [PubMed]
  53. Cowen, P.J. Serotonin and Depression: Pathophysiological Mechanism or Marketing Myth? Trends Pharmacol. Sci. 2008, 29, 433–436. [Google Scholar] [CrossRef]
  54. Albert, P.R.; Benkelfat, C.; Descarries, L. The Neurobiology of Depression--Revisiting the Serotonin Hypothesis. I. Cellular and Molecular Mechanisms. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2012, 367, 2378–2381. [Google Scholar] [CrossRef] [PubMed]
  55. Leyton, M.; Young, S.N.; Benkelfat, C. Relapse of Depression after Rapid Depletion of Tryptophan. Lancet Lond. Engl. 1997, 349, 1840–1841. [Google Scholar] [CrossRef]
  56. Delgado, P.L. Monoamine Depletion Studies: Implications for Antidepressant Discontinuation Syndrome. J. Clin. Psychiatry 2006, 67 (Suppl. 4), 22–26. [Google Scholar]
  57. Ślifirski, G.; Król, M.; Turło, J. 5-HT Receptors and the Development of New Antidepressants. Int. J. Mol. Sci. 2021, 22, 9015. [Google Scholar] [CrossRef]
  58. Orrico-Sanchez, A.; Chausset-Boissarie, L.; Alves De Sousa, R.; Coutens, B.; Rezai Amin, S.; Vialou, V.; Louis, F.; Hessani, A.; Dansette, P.M.; Zornoza, T.; et al. Antidepressant Efficacy of a Selective Organic Cation Transporter Blocker in a Mouse Model of Depression. Mol. Psychiatry 2020, 25, 1245–1259. [Google Scholar] [CrossRef]
  59. Spencer, R.L.; Chun, L.E.; Hartsock, M.J.; Woodruff, E.R. Glucocorticoid Hormones Are Both a Major Circadian Signal and Major Stress Signal: How This Shared Signal Contributes to a Dynamic Relationship between the Circadian and Stress Systems. Front. Neuroendocrinol. 2018, 49, 52–71. [Google Scholar] [CrossRef]
  60. Bunney, B.G.; Li, J.Z.; Walsh, D.M.; Stein, R.; Vawter, M.P.; Cartagena, P.; Barchas, J.D.; Schatzberg, A.F.; Myers, R.M.; Watson, S.J.; et al. Circadian Dysregulation of Clock Genes: Clues to Rapid Treatments in Major Depressive Disorder. Mol. Psychiatry 2015, 20, 48–55. [Google Scholar] [CrossRef]
  61. Kent, J.; Meredith, A.L. BK Channels Regulate Spontaneous Action Potential Rhythmicity in the Suprachiasmatic Nucleus. PLoS ONE 2008, 3, e3884. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, J.; Kim, D.E.; Griffin, P.; Sheehan, P.W.; Kim, D.-H.; Musiek, E.S.; Yoon, S.-Y. Inhibition of REV-ERBs Stimulates Microglial Amyloid-Beta Clearance and Reduces Amyloid Plaque Deposition in the 5XFAD Mouse Model of Alzheimer’s Disease. Aging Cell 2020, 19, e13078. [Google Scholar] [CrossRef] [PubMed]
  63. Zhu, Y.; Zhan, G.; Fenik, P.; Brandes, M.; Bell, P.; Francois, N.; Shulman, K.; Veasey, S. Chronic Sleep Disruption Advances the Temporal Progression of Tauopathy in P301S Mutant Mice. J. Neurosci. Off. J. Soc. Neurosci. 2018, 38, 10255–10270. [Google Scholar] [CrossRef] [PubMed]
  64. Daut, R.A.; Fonken, L.K. Circadian Regulation of Depression: A Role for Serotonin. Front. Neuroendocrinol. 2019, 54, 100746. [Google Scholar] [CrossRef]
  65. Kiss, J.; Léránth, C.; Halász, B. Serotoninergic Endings on VIP-Neurons in the Suprachiasmatic Nucleus and on ACTH-Neurons in the Arcuate Nucleus of the Rat Hypothalamus. A Combination of High Resolution Autoradiography and Electron Microscopic Immunocytochemistry. Neurosci. Lett. 1984, 44, 119–124. [Google Scholar] [CrossRef]
  66. Bosler, O.; Beaudet, A. VIP Neurons as Prime Synaptic Targets for Serotonin Afferents in Rat Suprachiasmatic Nucleus: A Combined Radioautographic and Immunocytochemical Study. J. Neurocytol. 1985, 14, 749–763. [Google Scholar] [CrossRef]
  67. Manrique, C.; Segu, L.; Héry, F.; Héry, M.; Faudon, M.; François-Bellan, A.M. Increase of Central 5-HT1B Binding Sites Following 5,7-Dihydroxytryptamine Axotomy in the Adult Rat. Brain Res. 1993, 623, 345–348. [Google Scholar] [CrossRef]
  68. Amir, S.; Robinson, B.; Ratovitski, T.; Rea, M.A.; Stewart, J.; Simantov, R. A Role for Serotonin in the Circadian System Revealed by the Distribution of Serotonin Transporter and Light-Induced Fos Immunoreactivity in the Suprachiasmatic Nucleus and Intergeniculate Leaflet. Neuroscience 1998, 84, 1059–1073. [Google Scholar] [CrossRef]
  69. Tao, L.; Jiang, R.; Zhang, K.; Qian, Z.; Chen, P.; Lv, Y.; Yao, Y. Light Therapy in Non-Seasonal Depression: An Update Meta-Analysis. Psychiatry Res. 2020, 291, 113247. [Google Scholar] [CrossRef]
  70. Richardson, C.R.; Faulkner, G.; McDevitt, J.; Skrinar, G.S.; Hutchinson, D.S.; Piette, J.D. Integrating Physical Activity into Mental Health Services for Persons with Serious Mental Illness. Psychiatr. Serv. Wash. DC 2005, 56, 324–331. [Google Scholar] [CrossRef]
  71. Hu, D.; Mao, Y.; Xu, G.; Liao, W.; Ren, J.; Yang, H.; Yang, J.; Sun, L.; Chen, H.; Wang, W.; et al. Time-Restricted Feeding Causes Irreversible Metabolic Disorders and Gut Microbiota Shift in Pediatric Mice. Pediatr. Res. 2019, 85, 518–526. [Google Scholar] [CrossRef] [PubMed]
  72. Upadhyay, A.; Anjum, B.; Godbole, N.M.; Rajak, S.; Shukla, P.; Tiwari, S.; Sinha, R.A.; Godbole, M.M. Time-Restricted Feeding Reduces High-Fat Diet Associated Placental Inflammation and Limits Adverse Effects on Fetal Organ Development. Biochem. Biophys. Res. Commun. 2019, 514, 415–421. [Google Scholar] [CrossRef]
  73. Yan, T.; Qiu, Y.; Yu, X.; Yang, L. Glymphatic Dysfunction: A Bridge Between Sleep Disturbance and Mood Disorders. Front. Psychiatry 2021, 12, 658340. [Google Scholar] [CrossRef] [PubMed]
  74. Reeves, B.C.; Karimy, J.K.; Kundishora, A.J.; Mestre, H.; Cerci, H.M.; Matouk, C.; Alper, S.L.; Lundgaard, I.; Nedergaard, M.; Kahle, K.T. Glymphatic System Impairment in Alzheimer’s Disease and Idiopathic Normal Pressure Hydrocephalus. Trends Mol. Med. 2020, 26, 285–295. [Google Scholar] [CrossRef] [PubMed]
  75. Shokri-Kojori, E.; Wang, G.-J.; Wiers, C.E.; Demiral, S.B.; Guo, M.; Kim, S.W.; Lindgren, E.; Ramirez, V.; Zehra, A.; Freeman, C.; et al. β-Amyloid Accumulation in the Human Brain after One Night of Sleep Deprivation. Proc. Natl. Acad. Sci. USA 2018, 115, 4483–4488. [Google Scholar] [CrossRef]
  76. López-Doménech, G.; Kittler, J.T. Mitochondrial Regulation of Local Supply of Energy in Neurons. Curr. Opin. Neurobiol. 2023, 81, 102747. [Google Scholar] [CrossRef]
  77. Alqahtani, T.; Deore, S.L.; Kide, A.A.; Shende, B.A.; Sharma, R.; Dadarao Chakole, R.; Nemade, L.S.; Kishor Kale, N.; Borah, S.; Shrikant Deokar, S.; et al. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease, and Parkinson’s Disease, Huntington’s Disease and Amyotrophic Lateral Sclerosis—An Updated Review. Mitochondrion 2023, 71, 83–92. [Google Scholar] [CrossRef]
  78. Cheng, X.-T.; Huang, N.; Sheng, Z.-H. Programming Axonal Mitochondrial Maintenance and Bioenergetics in Neurodegeneration and Regeneration. Neuron 2022, 110, 1899–1923. [Google Scholar] [CrossRef]
  79. Lambert, M.P.; Barlow, A.K.; Chromy, B.A.; Edwards, C.; Freed, R.; Liosatos, M.; Morgan, T.E.; Rozovsky, I.; Trommer, B.; Viola, K.L.; et al. Diffusible, Nonfibrillar Ligands Derived from Abeta1-42 Are Potent Central Nervous System Neurotoxins. Proc. Natl. Acad. Sci. USA 1998, 95, 6448–6453. [Google Scholar] [CrossRef]
  80. Cline, E.N.; Bicca, M.A.; Viola, K.L.; Klein, W.L. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J. Alzheimers Dis. JAD 2018, 64, S567–S610. [Google Scholar] [CrossRef]
  81. Bode, D.C.; Baker, M.D.; Viles, J.H. Ion Channel Formation by Amyloid-Β42 Oligomers but Not Amyloid-Β40 in Cellular Membranes. J. Biol. Chem. 2017, 292, 1404–1413. [Google Scholar] [CrossRef]
  82. Arispe, N. Architecture of the Alzheimer’s A Beta P Ion Channel Pore. J. Membr. Biol. 2004, 197, 33–48. [Google Scholar] [CrossRef] [PubMed]
  83. Sciacca, M.F.M.; Kotler, S.A.; Brender, J.R.; Chen, J.; Lee, D.; Ramamoorthy, A. Two-Step Mechanism of Membrane Disruption by Aβ through Membrane Fragmentation and Pore Formation. Biophys. J. 2012, 103, 702–710. [Google Scholar] [CrossRef] [PubMed]
  84. Kostylev, M.A.; Kaufman, A.C.; Nygaard, H.B.; Patel, P.; Haas, L.T.; Gunther, E.C.; Vortmeyer, A.; Strittmatter, S.M. Prion-Protein-Interacting Amyloid-β Oligomers of High Molecular Weight Are Tightly Correlated with Memory Impairment in Multiple Alzheimer Mouse Models. J. Biol. Chem. 2015, 290, 17415–17438. [Google Scholar] [CrossRef]
  85. Ohnishi, T.; Yanazawa, M.; Sasahara, T.; Kitamura, Y.; Hiroaki, H.; Fukazawa, Y.; Kii, I.; Nishiyama, T.; Kakita, A.; Takeda, H.; et al. Na, K-ATPase A3 Is a Death Target of Alzheimer Patient Amyloid-β Assembly. Proc. Natl. Acad. Sci. USA 2015, 112, E4465–E4474. [Google Scholar] [CrossRef]
  86. Um, J.W.; Nygaard, H.B.; Heiss, J.K.; Kostylev, M.A.; Stagi, M.; Vortmeyer, A.; Wisniewski, T.; Gunther, E.C.; Strittmatter, S.M. Alzheimer Amyloid-β Oligomer Bound to Postsynaptic Prion Protein Activates Fyn to Impair Neurons. Nat. Neurosci. 2012, 15, 1227–1235. [Google Scholar] [CrossRef]
  87. Lee, J.-H.; Yang, D.-S.; Goulbourne, C.N.; Im, E.; Stavrides, P.; Pensalfini, A.; Chan, H.; Bouchet-Marquis, C.; Bleiwas, C.; Berg, M.J.; et al. Faulty Autolysosome Acidification in Alzheimer’s Disease Mouse Models Induces Autophagic Build-up of Aβ in Neurons, Yielding Senile Plaques. Nat. Neurosci. 2022, 25, 688–701. [Google Scholar] [CrossRef] [PubMed]
  88. Schützmann, M.P.; Hasecke, F.; Bachmann, S.; Zielinski, M.; Hänsch, S.; Schröder, G.F.; Zempel, H.; Hoyer, W. Endo-Lysosomal Aβ Concentration and pH Trigger Formation of Aβ Oligomers That Potently Induce Tau Missorting. Nat. Commun. 2021, 12, 4634. [Google Scholar] [CrossRef]
  89. Sebollela, A.; Cline, E.N.; Popova, I.; Luo, K.; Sun, X.; Ahn, J.; Barcelos, M.A.; Bezerra, V.N.; Lyra E Silva, N.M.; Patel, J.; et al. A Human scFv Antibody That Targets and Neutralizes High Molecular Weight Pathogenic Amyloid-β Oligomers. J. Neurochem. 2017, 142, 934–947. [Google Scholar] [CrossRef]
  90. Viola, K.L.; Bicca, M.A.; Bebenek, A.M.; Kranz, D.L.; Nandwana, V.; Waters, E.A.; Haney, C.R.; Lee, M.; Gupta, A.; Brahmbhatt, Z.; et al. The Therapeutic and Diagnostic Potential of Amyloid β Oligomers Selective Antibodies to Treat Alzheimer’s Disease. Front. Neurosci. 2021, 15, 768646. [Google Scholar] [CrossRef]
  91. Baloyannis, S.J. Mitochondrial Alterations in Alzheimer’s Disease. J. Alzheimers Dis. 2006, 9, 119–126. [Google Scholar] [CrossRef] [PubMed]
  92. Krishnan, K.J.; Ratnaike, T.E.; De Gruyter, H.L.M.; Jaros, E.; Turnbull, D.M. Mitochondrial DNA Deletions Cause the Biochemical Defect Observed in Alzheimer’s Disease. Neurobiol. Aging 2012, 33, 2210–2214. [Google Scholar] [CrossRef]
  93. McKenna, M.C.; Stridh, M.H.; McNair, L.F.; Sonnewald, U.; Waagepetersen, H.S.; Schousboe, A. Glutamate Oxidation in Astrocytes: Roles of Glutamate Dehydrogenase and Aminotransferases. J. Neurosci. Res. 2016, 94, 1561–1571. [Google Scholar] [CrossRef] [PubMed]
  94. Arrázola, M.S.; Ramos-Fernández, E.; Cisternas, P.; Ordenes, D.; Inestrosa, N.C. Wnt Signaling Prevents the Aβ Oligomer-Induced Mitochondrial Permeability Transition Pore Opening Preserving Mitochondrial Structure in Hippocampal Neurons. PLoS ONE 2017, 12, e0168840. [Google Scholar] [CrossRef] [PubMed]
  95. Sorrentino, V.; Romani, M.; Mouchiroud, L.; Beck, J.S.; Zhang, H.; D’Amico, D.; Moullan, N.; Potenza, F.; Schmid, A.W.; Rietsch, S.; et al. Enhancing Mitochondrial Proteostasis Reduces Amyloid-β Proteotoxicity. Nature 2017, 552, 187–193. [Google Scholar] [CrossRef]
  96. Perez Ortiz, J.M.; Swerdlow, R.H. Mitochondrial Dysfunction in Alzheimer’s Disease: Role in Pathogenesis and Novel Therapeutic Opportunities. Br. J. Pharmacol. 2019, 176, 3489–3507. [Google Scholar] [CrossRef]
  97. Calvo-Rodriguez, M.; Hou, S.S.; Snyder, A.C.; Kharitonova, E.K.; Russ, A.N.; Das, S.; Fan, Z.; Muzikansky, A.; Garcia-Alloza, M.; Serrano-Pozo, A.; et al. Increased Mitochondrial Calcium Levels Associated with Neuronal Death in a Mouse Model of Alzheimer’s Disease. Nat. Commun. 2020, 11, 2146. [Google Scholar] [CrossRef]
  98. Sayyed, U.M.H.; Mahalakshmi, R. Mitochondrial Protein Translocation Machinery: From TOM Structural Biogenesis to Functional Regulation. J. Biol. Chem. 2022, 298, 101870. [Google Scholar] [CrossRef]
  99. Sirk, D.; Zhu, Z.; Wadia, J.S.; Shulyakova, N.; Phan, N.; Fong, J.; Mills, L.R. Chronic Exposure to Sub-Lethal Beta-Amyloid (Abeta) Inhibits the Import of Nuclear-Encoded Proteins to Mitochondria in Differentiated PC12 Cells. J. Neurochem. 2007, 103, 1989–2003. [Google Scholar] [CrossRef]
  100. Meng, X.; Song, Q.; Liu, Z.; Liu, X.; Wang, Y.; Liu, J. Neurotoxic β-Amyloid Oligomers Cause Mitochondrial Dysfunction-the Trigger for PANoptosis in Neurons. Front. Aging Neurosci. 2024, 16, 1400544. [Google Scholar] [CrossRef]
  101. Sahay, A.; Hen, R. Adult Hippocampal Neurogenesis in Depression. Nat. Neurosci. 2007, 10, 1110–1115. [Google Scholar] [CrossRef]
  102. Vaidya, V.A.; Fernandes, K.; Jha, S. Regulation of Adult Hippocampal Neurogenesis: Relevance to Depression. Expert Rev. Neurother. 2007, 7, 853–864. [Google Scholar] [CrossRef] [PubMed]
  103. Diksic, M. Labelled Alpha-Methyl-L-Tryptophan as a Tracer for the Study of the Brain Serotonergic System. J. Psychiatry Neurosci. JPN 2001, 26, 293–303. [Google Scholar] [PubMed]
  104. Albrecht, J.; Zielińska, M. Exchange-Mode Glutamine Transport across CNS Cell Membranes. Neuropharmacology 2019, 161, 107560. [Google Scholar] [CrossRef] [PubMed]
  105. Abela, A.R.; Browne, C.J.; Sargin, D.; Prevot, T.D.; Ji, X.D.; Li, Z.; Lambe, E.K.; Fletcher, P.J. Median Raphe Serotonin Neurons Promote Anxiety-like Behavior via Inputs to the Dorsal Hippocampus. Neuropharmacology 2020, 168, 107985. [Google Scholar] [CrossRef]
  106. Zhang, X.; Beaulieu, J.-M.; Sotnikova, T.D.; Gainetdinov, R.R.; Caron, M.G. Tryptophan Hydroxylase-2 Controls Brain Serotonin Synthesis. Science 2004, 305, 217. [Google Scholar] [CrossRef]
  107. Galano, A.; Castañeda-Arriaga, R.; Pérez-González, A.; Tan, D.-X.; Reiter, R.J. Phenolic Melatonin-Related Compounds: Their Role as Chemical Protectors against Oxidative Stress. Molecules 2016, 21, 1442. [Google Scholar] [CrossRef]
  108. Fanibunda, S.E.; Deb, S.; Maniyadath, B.; Tiwari, P.; Ghai, U.; Gupta, S.; Figueiredo, D.; Weisstaub, N.; Gingrich, J.A.; Vaidya, A.D.B.; et al. Serotonin Regulates Mitochondrial Biogenesis and Function in Rodent Cortical Neurons via the 5-HT 2A Receptor and SIRT1–PGC-1α Axis. Proc. Natl. Acad. Sci. USA 2019, 116, 11028–11037. [Google Scholar] [CrossRef]
  109. Azmitia, E.C. Serotonin and Brain: Evolution, Neuroplasticity, and Homeostasis. Int. Rev. Neurobiol. 2007, 77, 31–56. [Google Scholar] [CrossRef]
  110. Velasquez, J.C.; Goeden, N.; Bonnin, A. Placental Serotonin: Implications for the Developmental Effects of SSRIs and Maternal Depression. Front. Cell. Neurosci. 2013, 7, 47. [Google Scholar] [CrossRef]
  111. Chaouloff, F.; Berton, O.; Mormède, P. Serotonin and Stress. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 1999, 21, 28S–32S. [Google Scholar] [CrossRef]
  112. Dalva, M.B.; McClelland, A.C.; Kayser, M.S. Cell Adhesion Molecules: Signalling Functions at the Synapse. Nat. Rev. Neurosci. 2007, 8, 206–220. [Google Scholar] [CrossRef] [PubMed]
  113. Daubert, E.A.; Condron, B.G. Serotonin: A Regulator of Neuronal Morphology and Circuitry. Trends Neurosci. 2010, 33, 424–434. [Google Scholar] [CrossRef] [PubMed]
  114. Gaspar, P.; Cases, O.; Maroteaux, L. The Developmental Role of Serotonin: News from Mouse Molecular Genetics. Nat. Rev. Neurosci. 2003, 4, 1002–1012. [Google Scholar] [CrossRef]
  115. Jin, Y.; Dougherty, S.E.; Wood, K.; Sun, L.; Cudmore, R.H.; Abdalla, A.; Kannan, G.; Pletnikov, M.; Hashemi, P.; Linden, D.J. Regrowth of Serotonin Axons in the Adult Mouse Brain Following Injury. Neuron 2016, 91, 748–762. [Google Scholar] [CrossRef]
  116. Kajstura, T.J.; Dougherty, S.E.; Linden, D.J. Serotonin Axons in the Neocortex of the Adult Female Mouse Regrow after Traumatic Brain Injury. J. Neurosci. Res. 2018, 96, 512–526. [Google Scholar] [CrossRef] [PubMed]
  117. Zahrai, A.; Vahid-Ansari, F.; Daigle, M.; Albert, P.R. Fluoxetine-Induced Recovery of Serotonin and Norepinephrine Projections in a Mouse Model of Post-Stroke Depression. Transl. Psychiatry 2020, 10, 334. [Google Scholar] [CrossRef]
  118. Turlejski, K. Evolutionary Ancient Roles of Serotonin: Long-Lasting Regulation of Activity and Development. Acta Neurobiol. Exp. 1996, 56, 619–636. [Google Scholar] [CrossRef]
  119. Lesch, K.-P.; Waider, J. Serotonin in the Modulation of Neural Plasticity and Networks: Implications for Neurodevelopmental Disorders. Neuron 2012, 76, 175–191. [Google Scholar] [CrossRef]
  120. Brummelte, S.; Mc Glanaghy, E.; Bonnin, A.; Oberlander, T.F. Developmental Changes in Serotonin Signaling: Implications for Early Brain Function, Behavior and Adaptation. Neuroscience 2017, 342, 212–231. [Google Scholar] [CrossRef]
  121. Lupien, S.J. Brains under Stress. Can. J. Psychiatry Rev. Can. Psychiatr. 2009, 54, 4–5. [Google Scholar] [CrossRef] [PubMed]
  122. Buschiazzo, A.; Alzari, P.M. Structural Insights into Sialic Acid Enzymology. Curr. Opin. Chem. Biol. 2008, 12, 565–572. [Google Scholar] [CrossRef]
  123. Cummings, K.J.; Leiter, J.C. Take a Deep Breath and Wake up: The Protean Role of Serotonin Preventing Sudden Death in Infancy. Exp. Neurol. 2020, 326, 113165. [Google Scholar] [CrossRef] [PubMed]
  124. Paulus, E.V.; Mintz, E.M. Circadian Rhythms of Clock Gene Expression in the Cerebellum of Serotonin-Deficient Pet-1 Knockout Mice. Brain Res. 2016, 1630, 10–17. [Google Scholar] [CrossRef] [PubMed]
  125. Olivier, J.D.A.; Vinkers, C.H.; Olivier, B. The Role of the Serotonergic and GABA System in Translational Approaches in Drug Discovery for Anxiety Disorders. Front. Pharmacol. 2013, 4, 74. [Google Scholar] [CrossRef]
  126. De Deurwaerdère, P.; Di Giovanni, G. Serotonin in Health and Disease. Int. J. Mol. Sci. 2020, 21, 3500. [Google Scholar] [CrossRef]
  127. De Deurwaerdère, P.; Chagraoui, A.; Di Giovanni, G. Serotonin/Dopamine Interaction: Electrophysiological and Neurochemical Evidence. Prog. Brain Res. 2021, 261, 161–264. [Google Scholar] [CrossRef]
  128. Iovino, L.; Mutolo, D.; Cinelli, E.; Contini, M.; Pantaleo, T.; Bongianni, F. Breathing Stimulation Mediated by 5-HT1A and 5-HT3 Receptors within the preBötzinger Complex of the Adult Rabbit. Brain Res. 2019, 1704, 26–39. [Google Scholar] [CrossRef]
  129. Murphy, D.L.; Lerner, A.; Rudnick, G.; Lesch, K.-P. Serotonin Transporter: Gene, Genetic Disorders, and Pharmacogenetics. Mol. Interv. 2004, 4, 109–123. [Google Scholar] [CrossRef]
  130. Lin, Z.; Madras, B.K. Human Genetics and Pharmacology of Neurotransmitter Transporters. Handb. Exp. Pharmacol. 2006, 175, 327–371. [Google Scholar] [CrossRef]
  131. Kristensen, A.S.; Andersen, J.; Jørgensen, T.N.; Sørensen, L.; Eriksen, J.; Loland, C.J.; Strømgaard, K.; Gether, U. SLC6 Neurotransmitter Transporters: Structure, Function, and Regulation. Pharmacol. Rev. 2011, 63, 585–640. [Google Scholar] [CrossRef]
  132. Duan, H.; Wang, J. Selective Transport of Monoamine Neurotransmitters by Human Plasma Membrane Monoamine Transporter and Organic Cation Transporter 3. J. Pharmacol. Exp. Ther. 2010, 335, 743–753. [Google Scholar] [CrossRef]
  133. Bockaert, J.; Claeysen, S.; Bécamel, C.; Dumuis, A.; Marin, P. Neuronal 5-HT Metabotropic Receptors: Fine-Tuning of Their Structure, Signaling, and Roles in Synaptic Modulation. Cell Tissue Res. 2006, 326, 553–572. [Google Scholar] [CrossRef]
  134. Serotonin Receptors in Neurobiology; Chattopadhyay, A., Ed.; Frontiers in Neuroscience; CRC Press: Boca Raton, FL, USA, 2007; ISBN 978-0-8493-3977-6. [Google Scholar]
  135. Nadeev, A.D.; Zharkikh, I.L.; Avdonin, P.V.; Goncharov, N.V. Serotonin and its receptors in the cardiovascular system. Eksp. Klin. Farmakol. 2014, 77, 32–37. [Google Scholar] [PubMed]
  136. Wang, Y.; Tu, D.; Du, J.; Han, X.; Sun, Y.; Xu, Q.; Zhai, G.; Zhou, Y. Classification of Subcortical Vascular Cognitive Impairment Using Single MRI Sequence and Deep Learning Convolutional Neural Networks. Front. Neurosci. 2019, 13, 627. [Google Scholar] [CrossRef] [PubMed]
  137. Noda, M.; Higashida, H.; Aoki, S.; Wada, K. Multiple Signal Transduction Pathways Mediated by 5-HT Receptors. Mol. Neurobiol. 2004, 29, 31–40. [Google Scholar] [CrossRef]
  138. Asada, M.; Ebihara, S.; Yamanda, S.; Niu, K.; Okazaki, T.; Sora, I.; Arai, H. Depletion of Serotonin and Selective Inhibition of 2B Receptor Suppressed Tumor Angiogenesis by Inhibiting Endothelial Nitric Oxide Synthase and Extracellular Signal-Regulated Kinase 1/2 Phosphorylation. Neoplasia 2009, 11, 408–417. [Google Scholar] [CrossRef] [PubMed]
  139. Charnay, Y.; Léger, L. Brain Serotonergic Circuitries. Dialogues Clin. Neurosci. 2010, 12, 471–487. [Google Scholar] [CrossRef]
  140. Davis, R.P.; Pattison, J.; Thompson, J.M.; Tiniakov, R.; Scrogin, K.E.; Watts, S.W. 5-Hydroxytryptamine (5-HT) Reduces Total Peripheral Resistance during Chronic Infusion: Direct Arterial Mesenteric Relaxation Is Not Involved. BMC Pharmacol. 2012, 12, 4. [Google Scholar] [CrossRef]
  141. Fujita, M.; Minamino, T.; Sanada, S.; Asanuma, H.; Hirata, A.; Ogita, H.; Okada, K.; Tsukamoto, O.; Takashima, S.; Tomoike, H.; et al. Selective Blockade of Serotonin 5-HT2A Receptor Increases Coronary Blood Flow via Augmented Cardiac Nitric Oxide Release through 5-HT1B Receptor in Hypoperfused Canine Hearts. J. Mol. Cell. Cardiol. 2004, 37, 1219–1223. [Google Scholar] [CrossRef]
  142. Liu, Y.; Fanburg, B.L. Serotonin-Induced Growth of Pulmonary Artery Smooth Muscle Requires Activation of Phosphatidylinositol 3-Kinase/Serine-Threonine Protein Kinase B/Mammalian Target of Rapamycin/P70 Ribosomal S6 Kinase 1. Am. J. Respir. Cell Mol. Biol. 2006, 34, 182–191. [Google Scholar] [CrossRef] [PubMed]
  143. Lee, S.L.; Wang, W.W.; Fanburg, B.L. Superoxide as an Intermediate Signal for Serotonin-Induced Mitogenesis. Free Radic. Biol. Med. 1998, 24, 855–858. [Google Scholar] [CrossRef] [PubMed]
  144. Kamata, H.; Honda, S.-I.; Maeda, S.; Chang, L.; Hirata, H.; Karin, M. Reactive Oxygen Species Promote TNFalpha-Induced Death and Sustained JNK Activation by Inhibiting MAP Kinase Phosphatases. Cell 2005, 120, 649–661. [Google Scholar] [CrossRef]
  145. Linder, A.E.; Gaskell, G.L.; Szasz, T.; Thompson, J.M.; Watts, S.W. Serotonin Receptors in Rat Jugular Vein: Presence and Involvement in the Contraction. J. Pharmacol. Exp. Ther. 2010, 334, 116–123. [Google Scholar] [CrossRef]
  146. Lu, R.; Alioua, A.; Kumar, Y.; Kundu, P.; Eghbali, M.; Weisstaub, N.V.; Gingrich, J.A.; Stefani, E.; Toro, L. C-Src Tyrosine Kinase, a Critical Component for 5-HT2A Receptor-Mediated Contraction in Rat Aorta. J. Physiol. 2008, 586, 3855–3869. [Google Scholar] [CrossRef] [PubMed]
  147. MacLean, M.M.R. The Serotonin Hypothesis in Pulmonary Hypertension Revisited: Targets for Novel Therapies (2017 Grover Conference Series). Pulm. Circ. 2018, 8, 2045894018759125. [Google Scholar] [CrossRef] [PubMed]
  148. Mukhin, Y.V.; Garnovskaya, M.N.; Collinsworth, G.; Grewal, J.S.; Pendergrass, D.; Nagai, T.; Pinckney, S.; Greene, E.L.; Raymond, J.R. 5-Hydroxytryptamine1A Receptor/Gibetagamma Stimulates Mitogen-Activated Protein Kinase via NAD(P)H Oxidase and Reactive Oxygen Species Upstream of Src in Chinese Hamster Ovary Fibroblasts. Biochem. J. 2000, 347 Pt 1, 61–67. [Google Scholar] [CrossRef]
  149. Nelson, D.L. 5-HT5 Receptors. Curr. Drug Targets CNS Neurol. Disord. 2004, 3, 53–58. [Google Scholar] [CrossRef]
  150. Nichols, D.E.; Nichols, C.D. Serotonin Receptors. Chem. Rev. 2008, 108, 1614–1641. [Google Scholar] [CrossRef]
  151. Ogden, K.; Thompson, J.M.; Hickner, Z.; Huang, T.; Tang, D.D.; Watts, S.W. A New Signaling Paradigm for Serotonin: Use of Crk-Associated Substrate in Arterial Contraction. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H2857–H2863. [Google Scholar] [CrossRef] [PubMed]
  152. Ullmer, C.; Boddeke, H.G.; Schmuck, K.; Lübbert, H. 5-HT2B Receptor-Mediated Calcium Release from Ryanodine-Sensitive Intracellular Stores in Human Pulmonary Artery Endothelial Cells. Br. J. Pharmacol. 1996, 117, 1081–1088. [Google Scholar] [CrossRef]
  153. Villalón, C.M.; Centurión, D. Cardiovascular Responses Produced by 5-Hydroxytriptamine:A Pharmacological Update on the Receptors/Mechanisms Involved and Therapeutic Implications. Naunyn. Schmiedebergs Arch. Pharmacol. 2007, 376, 45–63. [Google Scholar] [CrossRef]
  154. Watts, S.W. 5-HT in Systemic Hypertension: Foe, Friend or Fantasy? Clin. Sci. Lond. Engl. 1979 2005, 108, 399–412. [Google Scholar] [CrossRef] [PubMed]
  155. Wei, L.; Liu, Y.; Kaneto, H.; Fanburg, B.L. JNK Regulates Serotonin-Mediated Proliferation and Migration of Pulmonary Artery Smooth Muscle Cells. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2010, 298, L863–L869. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, Y.; Zhang, L.; Yu, J.; Ma, Z.; Li, M.; Wang, J.; Hu, P.; Zou, J.; Liu, X.; Liu, Y.; et al. A Novel 5-HT1B Receptor Agonist of Herbal Compounds and One of the Therapeutic Uses for Alzheimer’s Disease. Front. Pharmacol. 2021, 12, 735876. [Google Scholar] [CrossRef] [PubMed]
  157. Fu, W.; Xie, H.; Laudon, M.; Zhou, S.; Tian, S.; You, Y. Piromelatine Ameliorates Memory Deficits Associated with Chronic Mild Stress-Induced Anhedonia in Rats. Psychopharmacology 2016, 233, 2229–2239. [Google Scholar] [CrossRef]
  158. Acquarone, E.; Argyrousi, E.K.; Arancio, O.; Watterson, D.M.; Roy, S.M. The 5HT2b Receptor in Alzheimer’s Disease: Increased Levels in Patient Brains and Antagonist Attenuation of Amyloid and Tau Induced Dysfunction. J. Alzheimers Dis. JAD 2024, 98, 1349–1360. [Google Scholar] [CrossRef]
  159. Liu, H.; He, Y.; Liu, H.; Brouwers, B.; Yin, N.; Lawler, K.; Keogh, J.M.; Henning, E.; Lee, D.-K.; Yu, M.; et al. Neural Circuits Expressing the Serotonin 2C Receptor Regulate Memory in Mice and Humans. Sci. Adv. 2024, 10, eadl2675. [Google Scholar] [CrossRef]
  160. Tian, Z.R.; Sharma, A.; Muresanu, D.F.; Sharma, S.; Feng, L.; Zhang, Z.; Li, C.; Buzoianu, A.D.; Lafuente, J.V.; Nozari, A.; et al. Nicotine Neurotoxicity Exacerbation Following Engineered Ag and Cu (50–60 Nm) Nanoparticles Intoxication. Neuroprotection with Nanowired Delivery of Antioxidant Compound H-290/51 Together with Serotonin 5-HT3 Receptor Antagonist Ondansetron. Int. Rev. Neurobiol. 2023, 172, 189–233. [Google Scholar] [CrossRef]
  161. Jiang, S.; Sydney, E.J.; Runyan, A.M.; Serpe, R.; Srikanth, M.; Figueroa, H.Y.; Yang, M.; Myeku, N. 5-HT4 Receptor Agonists Treatment Reduces Tau Pathology and Behavioral Deficit in the PS19 Mouse Model of Tauopathy. Front. Cell. Neurosci. 2024, 18, 1338502. [Google Scholar] [CrossRef]
  162. Wu, J.; Li, Q.; Bezprozvanny, I. Evaluation of Dimebon in Cellular Model of Huntington’s Disease. Mol. Neurodegener. 2008, 3, 15. [Google Scholar] [CrossRef] [PubMed]
  163. Chaumont-Dubel, S.; Dupuy, V.; Bockaert, J.; Bécamel, C.; Marin, P. The 5-HT6 Receptor Interactome: New Insight in Receptor Signaling and Its Impact on Brain Physiology and Pathologies. Neuropharmacology 2020, 172, 107839. [Google Scholar] [CrossRef]
  164. Quintero-Villegas, A.; Valdés-Ferrer, S.I. Central Nervous System Effects of 5-HT7 Receptors: A Potential Target for Neurodegenerative Diseases. Mol. Med. Camb. Mass 2022, 28, 70. [Google Scholar] [CrossRef]
  165. McDevitt, R.A.; Neumaier, J.F. Regulation of Dorsal Raphe Nucleus Function by Serotonin Autoreceptors: A Behavioral Perspective. J. Chem. Neuroanat. 2011, 41, 234–246. [Google Scholar] [CrossRef] [PubMed]
  166. Courtney, N.A.; Ford, C.P. Mechanisms of 5-HT1A Receptor-Mediated Transmission in Dorsal Raphe Serotonin Neurons. J. Physiol. 2016, 594, 953–965. [Google Scholar] [CrossRef]
  167. Llamosas, N.; Bruzos-Cidón, C.; Rodríguez, J.J.; Ugedo, L.; Torrecilla, M. Deletion of GIRK2 Subunit of GIRK Channels Alters the 5-HT 1A Receptor-Mediated Signaling and Results in a Depression-Resistant Behavior. Int. J. Neuropsychopharmacol. 2015, 18, pyv051. [Google Scholar] [CrossRef] [PubMed]
  168. Montalbano, A.; Corradetti, R.; Mlinar, B. Pharmacological Characterization of 5-HT1A Autoreceptor-Coupled GIRK Channels in Rat Dorsal Raphe 5-HT Neurons. PLoS ONE 2015, 10, e0140369. [Google Scholar] [CrossRef]
  169. Grunnet, M.; Jespersen, T.; Perrier, J.-F. 5-HT1A Receptors Modulate Small-Conductance Ca2+-Activated K+ Channels. J. Neurosci. Res. 2004, 78, 845–854. [Google Scholar] [CrossRef]
  170. Kirby, L.G.; Pernar, L.; Valentino, R.J.; Beck, S.G. Distinguishing Characteristics of Serotonin and Non-Serotonin-Containing Cells in the Dorsal Raphe Nucleus: Electrophysiological and Immunohistochemical Studies. Neuroscience 2003, 116, 669–683. [Google Scholar] [CrossRef]
  171. Chen, S.; Owens, G.C.; Crossin, K.L.; Edelman, D.B. Serotonin Stimulates Mitochondrial Transport in Hippocampal Neurons. Mol. Cell. Neurosci. 2007, 36, 472–483. [Google Scholar] [CrossRef]
  172. Albert, P.R.; Lemonde, S. 5-HT1A Receptors, Gene Repression, and Depression: Guilt by Association. Neurosci. Rev. J. Bringing Neurobiol. Neurol. Psychiatry 2004, 10, 575–593. [Google Scholar] [CrossRef] [PubMed]
  173. Altieri, S.C.; Garcia-Garcia, A.L.; Leonardo, E.D.; Andrews, A.M. Rethinking 5-HT1A Receptors: Emerging Modes of Inhibitory Feedback of Relevance to Emotion-Related Behavior. ACS Chem. Neurosci. 2013, 4, 72–83. [Google Scholar] [CrossRef]
  174. Vargas, M.V.; Dunlap, L.E.; Dong, C.; Carter, S.J.; Tombari, R.J.; Jami, S.A.; Cameron, L.P.; Patel, S.D.; Hennessey, J.J.; Saeger, H.N.; et al. Psychedelics Promote Neuroplasticity through the Activation of Intracellular 5-HT2A Receptors. Science 2023, 379, 700–706. [Google Scholar] [CrossRef]
  175. Hornung, J.-P. The Human Raphe Nuclei and the Serotonergic System. J. Chem. Neuroanat. 2003, 26, 331–343. [Google Scholar] [CrossRef]
  176. Carhart-Harris, R.L. Serotonin, Psychedelics and Psychiatry. World Psychiatry Off. J. World Psychiatr. Assoc. WPA 2018, 17, 358–359. [Google Scholar] [CrossRef]
  177. Aleksandrova, L.R.; Phillips, A.G. Neuroplasticity as a Convergent Mechanism of Ketamine and Classical Psychedelics. Trends Pharmacol. Sci. 2021, 42, 929–942. [Google Scholar] [CrossRef] [PubMed]
  178. Wallach, J.; Cao, A.B.; Calkins, M.M.; Heim, A.J.; Lanham, J.K.; Bonniwell, E.M.; Hennessey, J.J.; Bock, H.A.; Anderson, E.I.; Sherwood, A.M.; et al. Identification of 5-HT2A Receptor Signaling Pathways Associated with Psychedelic Potential. Nat. Commun. 2023, 14, 8221. [Google Scholar] [CrossRef]
  179. Cornea-Hébert, V.; Watkins, K.C.; Roth, B.L.; Kroeze, W.K.; Gaudreau, P.; Leclerc, N.; Descarries, L. Similar Ultrastructural Distribution of the 5-HT(2A) Serotonin Receptor and Microtubule-Associated Protein MAP1A in Cortical Dendrites of Adult Rat. Neuroscience 2002, 113, 23–35. [Google Scholar] [CrossRef] [PubMed]
  180. Béïque, J.-C.; Imad, M.; Mladenovic, L.; Gingrich, J.A.; Andrade, R. Mechanism of the 5-Hydroxytryptamine 2A Receptor-Mediated Facilitation of Synaptic Activity in Prefrontal Cortex. Proc. Natl. Acad. Sci. USA 2007, 104, 9870–9875. [Google Scholar] [CrossRef]
  181. Andrade, R. Serotonergic Regulation of Neuronal Excitability in the Prefrontal Cortex. Neuropharmacology 2011, 61, 382–386. [Google Scholar] [CrossRef]
  182. Aznar, S.; Hervig, M.E.-S. The 5-HT2A Serotonin Receptor in Executive Function: Implications for Neuropsychiatric and Neurodegenerative Diseases. Neurosci. Biobehav. Rev. 2016, 64, 63–82. [Google Scholar] [CrossRef] [PubMed]
  183. Kwan, A.C.; Olson, D.E.; Preller, K.H.; Roth, B.L. The Neural Basis of Psychedelic Action. Nat. Neurosci. 2022, 25, 1407–1419. [Google Scholar] [CrossRef] [PubMed]
  184. Yakel, J.L.; Shao, X.M.; Jackson, M.B. The Selectivity of the Channel Coupled to the 5-HT3 Receptor. Brain Res. 1990, 533, 46–52. [Google Scholar] [CrossRef]
  185. Malone, H.M.; Peters, J.A.; Lambert, J.J. Physiological and Pharmacological Properties of 5-HT3 Receptors--a Patch Clamp-Study. Neuropeptides 1991, 19, 25–30. [Google Scholar] [CrossRef]
  186. Maricq, A.V.; Peterson, A.S.; Brake, A.J.; Myers, R.M.; Julius, D. Primary Structure and Functional Expression of the 5HT3 Receptor, a Serotonin-Gated Ion Channel. Science 1991, 254, 432–437. [Google Scholar] [CrossRef]
  187. Niesler, B.; Frank, B.; Kapeller, J.; Rappold, G.A. Cloning, Physical Mapping and Expression Analysis of the Human 5-HT3 Serotonin Receptor-like Genes HTR3C, HTR3D and HTR3E. Gene 2003, 310, 101–111. [Google Scholar] [CrossRef] [PubMed]
  188. Blandina, P.; Goldfarb, J.; Craddock-Royal, B.; Green, J.P. Release of Endogenous Dopamine by Stimulation of 5-Hydroxytryptamine3 Receptors in Rat Striatum. J. Pharmacol. Exp. Ther. 1989, 251, 803–809. [Google Scholar] [CrossRef]
  189. Miquel, M.-C.; Emerit, M.B.; Nosjean, A.; Simon, A.; Rumajogee, P.; Brisorgueil, M.-J.; Doucet, E.; Hamon, M.; Vergé, D. Differential Subcellular Localization of the 5-HT3-As Receptor Subunit in the Rat Central Nervous System. Eur. J. Neurosci. 2002, 15, 449–457. [Google Scholar] [CrossRef]
  190. Thompson, A.J.; Lummis, S.C.R. 5-HT3 Receptors. Curr. Pharm. Des. 2006, 12, 3615–3630. [Google Scholar] [CrossRef]
  191. Bloom, F.E.; Morales, M. The Central 5-HT3 Receptor in CNS Disorders. Neurochem. Res. 1998, 23, 653–659. [Google Scholar] [CrossRef]
  192. Morales, M.; Bloom, F.E. The 5-HT3 Receptor Is Present in Different Subpopulations of GABAergic Neurons in the Rat Telencephalon. J. Neurosci. Off. J. Soc. Neurosci. 1997, 17, 3157–3167. [Google Scholar] [CrossRef]
  193. Turner, T.J.; Mokler, D.J.; Luebke, J.I. Calcium Influx through Presynaptic 5-HT3 Receptors Facilitates GABA Release in the Hippocampus: In Vitro Slice and Synaptosome Studies. Neuroscience 2004, 129, 703–718. [Google Scholar] [CrossRef] [PubMed]
  194. Krauthausen, M.; Kummer, M.P.; Zimmermann, J.; Reyes-Irisarri, E.; Terwel, D.; Bulic, B.; Heneka, M.T.; Müller, M. CXCR3 Promotes Plaque Formation and Behavioral Deficits in an Alzheimer’s Disease Model. J. Clin. Investig. 2015, 125, 365–378. [Google Scholar] [CrossRef]
  195. Liu, L.-F.; Liu, Y.-T.; Wu, D.-D.; Cheng, J.; Li, N.-N.; Zheng, Y.-N.; Huang, L.; Yuan, Q.-L. Inhibiting 5-Hydroxytryptamine Receptor 3 Alleviates Pathological Changes of a Mouse Model of Alzheimer’s Disease. Neural Regen. Res. 2023, 18, 2019–2028. [Google Scholar] [CrossRef] [PubMed]
  196. Bockaert, J.; Claeysen, S.; Compan, V.; Dumuis, A. 5-HT(4) Receptors: History, Molecular Pharmacology and Brain Functions. Neuropharmacology 2008, 55, 922–931. [Google Scholar] [CrossRef] [PubMed]
  197. Bonaventure, P.; Hall, H.; Gommeren, W.; Cras, P.; Langlois, X.; Jurzak, M.; Leysen, J.E. Mapping of Serotonin 5-HT(4) Receptor mRNA and Ligand Binding Sites in the Post-Mortem Human Brain. Synapse 2000, 36, 35–46. [Google Scholar] [CrossRef]
  198. Karayol, R.; Medrihan, L.; Warner-Schmidt, J.L.; Fait, B.W.; Rao, M.N.; Holzner, E.B.; Greengard, P.; Heintz, N.; Schmidt, E.F. Serotonin Receptor 4 in the Hippocampus Modulates Mood and Anxiety. Mol. Psychiatry 2021, 26, 2334–2349. [Google Scholar] [CrossRef]
  199. Lucas, G.; Rymar, V.V.; Du, J.; Mnie-Filali, O.; Bisgaard, C.; Manta, S.; Lambas-Senas, L.; Wiborg, O.; Haddjeri, N.; Piñeyro, G.; et al. Serotonin(4) (5-HT(4)) Receptor Agonists Are Putative Antidepressants with a Rapid Onset of Action. Neuron 2007, 55, 712–725. [Google Scholar] [CrossRef]
  200. Faye, C.; Hen, R.; Guiard, B.P.; Denny, C.A.; Gardier, A.M.; Mendez-David, I.; David, D.J. Rapid Anxiolytic Effects of RS67333, a Serotonin Type 4 Receptor Agonist, and Diazepam, a Benzodiazepine, Are Mediated by Projections From the Prefrontal Cortex to the Dorsal Raphe Nucleus. Biol. Psychiatry 2020, 87, 514–525. [Google Scholar] [CrossRef]
  201. Peñas-Cazorla, R.; Vilaró, M.T. Serotonin 5-HT4 Receptors and Forebrain Cholinergic System: Receptor Expression in Identified Cell Populations. Brain Struct. Funct. 2015, 220, 3413–3434. [Google Scholar] [CrossRef]
  202. Köhler-Forsberg, K.; Dam, V.H.; Ozenne, B.; Sankar, A.; Beliveau, V.; Landman, E.B.; Larsen, S.V.; Poulsen, A.S.; Ip, C.-T.; Jørgensen, A.; et al. Serotonin 4 Receptor Brain Binding in Major Depressive Disorder and Association With Memory Dysfunction. JAMA Psychiatry 2023, 80, 296–304. [Google Scholar] [CrossRef] [PubMed]
  203. Thomas, D.R. 5-ht5A Receptors as a Therapeutic Target. Pharmacol. Ther. 2006, 111, 707–714. [Google Scholar] [CrossRef]
  204. Vidal-Cantú, G.C.; Jiménez-Hernández, M.; Rocha-González, H.I.; Villalón, C.M.; Granados-Soto, V.; Muñoz-Islas, E. Role of 5-HT5A and 5-HT1B/1D Receptors in the Antinociception Produced by Ergotamine and Valerenic Acid in the Rat Formalin Test. Eur. J. Pharmacol. 2016, 781, 109–116. [Google Scholar] [CrossRef]
  205. Yun, H.-M.; Kim, S.; Kim, H.-J.; Kostenis, E.; Kim, J.I.; Seong, J.Y.; Baik, J.-H.; Rhim, H. The Novel Cellular Mechanism of Human 5-HT6 Receptor through an Interaction with Fyn. J. Biol. Chem. 2007, 282, 5496–5505. [Google Scholar] [CrossRef] [PubMed]
  206. Yun, H.-M.; Baik, J.-H.; Kang, I.; Jin, C.; Rhim, H. Physical Interaction of Jab1 with Human Serotonin 6 G-Protein-Coupled Receptor and Their Possible Roles in Cell Survival. J. Biol. Chem. 2010, 285, 10016–10029. [Google Scholar] [CrossRef] [PubMed]
  207. Riccioni, T.; Bordi, F.; Minetti, P.; Spadoni, G.; Yun, H.-M.; Im, B.-H.; Tarzia, G.; Rhim, H.; Borsini, F. ST1936 Stimulates cAMP, Ca2+, ERK1/2 and Fyn Kinase through a Full Activation of Cloned Human 5-HT6 Receptors. Eur. J. Pharmacol. 2011, 661, 8–14. [Google Scholar] [CrossRef]
  208. Riccio, O.; Potter, G.; Walzer, C.; Vallet, P.; Szabó, G.; Vutskits, L.; Kiss, J.Z.; Dayer, A.G. Excess of Serotonin Affects Embryonic Interneuron Migration through Activation of the Serotonin Receptor 6. Mol. Psychiatry 2009, 14, 280–290. [Google Scholar] [CrossRef]
  209. Jacobshagen, M.; Niquille, M.; Chaumont-Dubel, S.; Marin, P.; Dayer, A. The Serotonin 6 Receptor Controls Neuronal Migration during Corticogenesis via a Ligand-Independent Cdk5-Dependent Mechanism. Dev. Camb. Engl. 2014, 141, 3370–3377. [Google Scholar] [CrossRef]
  210. Guadiana, S.M.; Semple-Rowland, S.; Daroszewski, D.; Madorsky, I.; Breunig, J.J.; Mykytyn, K.; Sarkisian, M.R. Arborization of Dendrites by Developing Neocortical Neurons Is Dependent on Primary Cilia and Type 3 Adenylyl Cyclase. J. Neurosci. Off. J. Soc. Neurosci. 2013, 33, 2626–2638. [Google Scholar] [CrossRef]
  211. Lesiak, A.J.; Brodsky, M.; Cohenca, N.; Croicu, A.G.; Neumaier, J.F. Restoration of Physiological Expression of 5-HT 6 Receptor into the Primary Cilia of Null Mutant Neurons Lengthens Both Primary Cilia and Dendrites. Mol. Pharmacol. 2018, 94, 731–742. [Google Scholar] [CrossRef]
  212. Roberts, J.C.; Reavill, C.; East, S.Z.; Harrison, P.J.; Patel, S.; Routledge, C.; Leslie, R.A. The Distribution of 5-HT6 Receptors in Rat Brain: An Autoradiographic Binding Study Using the Radiolabelled 5-HT6 Receptor Antagonist [125I]SB-258585. Brain Res. 2002, 934, 49–57. [Google Scholar] [CrossRef] [PubMed]
  213. Woolley, M.L.; Marsden, C.A.; Fone, K.C.F. 5-Ht6 Receptors. Curr. Drug Targets CNS Neurol. Disord. 2004, 3, 59–79. [Google Scholar] [CrossRef] [PubMed]
  214. Helboe, L.; Egebjerg, J.; de Jong, I.E.M. Distribution of Serotonin Receptor 5-HT6 mRNA in Rat Neuronal Subpopulations: A Double in Situ Hybridization Study. Neuroscience 2015, 310, 442–454. [Google Scholar] [CrossRef] [PubMed]
  215. Dupuy, V.; Prieur, M.; Pizzoccaro, A.; Margarido, C.; Valjent, E.; Bockaert, J.; Bouschet, T.; Marin, P.; Chaumont-Dubel, S. Spatiotemporal Dynamics of 5-HT6 Receptor Ciliary Localization during Mouse Brain Development. Neurobiol. Dis. 2023, 176, 105949. [Google Scholar] [CrossRef]
  216. Meffre, J.; Chaumont-Dubel, S.; Mannoury La Cour, C.; Loiseau, F.; Watson, D.J.G.; Dekeyne, A.; Séveno, M.; Rivet, J.; Gaven, F.; Déléris, P.; et al. 5-HT 6 Receptor Recruitment of mTOR as a Mechanism for Perturbed Cognition in Schizophrenia. EMBO Mol. Med. 2012, 4, 1043–1056. [Google Scholar] [CrossRef]
  217. Garcia-Alloza, M.; Hirst, W.D.; Chen, C.P.L.-H.; Lasheras, B.; Francis, P.T.; Ramírez, M.J. Differential Involvement of 5-HT(1B/1D) and 5-HT6 Receptors in Cognitive and Non-Cognitive Symptoms in Alzheimer’s Disease. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2004, 29, 410–416. [Google Scholar] [CrossRef]
  218. Upton, N.; Chuang, T.T.; Hunter, A.J.; Virley, D.J. 5-HT6 Receptor Antagonists as Novel Cognitive Enhancing Agents for Alzheimer’s Disease. Neurother. J. Am. Soc. Exp. Neurother. 2008, 5, 458–469. [Google Scholar] [CrossRef]
  219. Foley, A.G.; Murphy, K.J.; Hirst, W.D.; Gallagher, H.C.; Hagan, J.J.; Upton, N.; Walsh, F.S.; Regan, C.M. The 5-HT(6) Receptor Antagonist SB-271046 Reverses Scopolamine-Disrupted Consolidation of a Passive Avoidance Task and Ameliorates Spatial Task Deficits in Aged Rats. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2004, 29, 93–100. [Google Scholar] [CrossRef]
  220. Benhamú, B.; Martín-Fontecha, M.; Vázquez-Villa, H.; Pardo, L.; López-Rodríguez, M.L. Serotonin 5-HT6 Receptor Antagonists for the Treatment of Cognitive Deficiency in Alzheimer’s Disease. J. Med. Chem. 2014, 57, 7160–7181. [Google Scholar] [CrossRef]
  221. Ivachtchenko, A.V.; Ivanenkov, Y.A.; Veselov, M.S.; Okun, I.M. AVN-322 Is a Safe Orally Bio-Available Potent and Highly Selective Antagonist of 5-HT6R with Demonstrated Ability to Improve Impaired Memory in Animal Models. Curr. Alzheimer Res. 2017, 14, 268–294. [Google Scholar] [CrossRef]
  222. Suárez-Santiago, J.E.; Roldán Roldán, G.; Picazo Picazo, O. The 5-HT6R Agonist E-6837 and the Antagonist SB-271046 Reverse the Psychotic-like Behaviors Induced by Ketamine. Behav. Pharmacol. 2022, 33, 249–254. [Google Scholar] [CrossRef]
  223. Kucwaj-Brysz, K.; Baltrukevich, H.; Czarnota, K.; Handzlik, J. Chemical Update on the Potential for Serotonin 5-HT6 and 5-HT7 Receptor Agents in the Treatment of Alzheimer’s Disease. Bioorg. Med. Chem. Lett. 2021, 49, 128275. [Google Scholar] [CrossRef] [PubMed]
  224. Heidmann, D.E.; Metcalf, M.A.; Kohen, R.; Hamblin, M.W. Four 5-Hydroxytryptamine7 (5-HT7) Receptor Isoforms in Human and Rat Produced by Alternative Splicing: Species Differences Due to Altered Intron-Exon Organization. J. Neurochem. 1997, 68, 1372–1381. [Google Scholar] [CrossRef] [PubMed]
  225. Adham, N.; Zgombick, J.M.; Bard, J.; Branchek, T.A. Functional Characterization of the Recombinant Human 5-Hydroxytryptamine7(a) Receptor Isoform Coupled to Adenylate Cyclase Stimulation. J. Pharmacol. Exp. Ther. 1998, 287, 508–514. [Google Scholar] [CrossRef] [PubMed]
  226. Guseva, D.; Wirth, A.; Ponimaskin, E. Cellular Mechanisms of the 5-HT7 Receptor-Mediated Signaling. Front. Behav. Neurosci. 2014, 8, 306. [Google Scholar] [CrossRef]
  227. Kobe, F.; Guseva, D.; Jensen, T.P.; Wirth, A.; Renner, U.; Hess, D.; Müller, M.; Medrihan, L.; Zhang, W.; Zhang, M.; et al. 5-HT7R/G12 Signaling Regulates Neuronal Morphology and Function in an Age-Dependent Manner. J. Neurosci. Off. J. Soc. Neurosci. 2012, 32, 2915–2930. [Google Scholar] [CrossRef]
  228. Speranza, L.; Giuliano, T.; Volpicelli, F.; De Stefano, M.E.; Lombardi, L.; Chambery, A.; Lacivita, E.; Leopoldo, M.; Bellenchi, G.C.; Di Porzio, U.; et al. Activation of 5-HT7 Receptor Stimulates Neurite Elongation through mTOR, Cdc42 and Actin Filaments Dynamics. Front. Behav. Neurosci. 2015, 9, 62. [Google Scholar] [CrossRef]
  229. Marin, P.; Dityatev, A. 5-HT7 Receptor Shapes Spinogenesis in Cortical and Striatal Neurons: An Editorial Highlight for “Serotonin 5-HT7 Receptor Increases the Density of Dendritic Spines and Facilitates Synaptogenesis in Forebrain Neurons”. J. Neurochem. 2017, 141, 644–646. [Google Scholar] [CrossRef]
  230. Speranza, L.; Chambery, A.; Di Domenico, M.; Crispino, M.; Severino, V.; Volpicelli, F.; Leopoldo, M.; Bellenchi, G.C.; di Porzio, U.; Perrone-Capano, C. The Serotonin Receptor 7 Promotes Neurite Outgrowth via ERK and Cdk5 Signaling Pathways. Neuropharmacology 2013, 67, 155–167. [Google Scholar] [CrossRef]
  231. Kvachnina, E.; Liu, G.; Dityatev, A.; Renner, U.; Dumuis, A.; Richter, D.W.; Dityateva, G.; Schachner, M.; Voyno-Yasenetskaya, T.A.; Ponimaskin, E.G. 5-HT7 Receptor Is Coupled to G Alpha Subunits of Heterotrimeric G12-Protein to Regulate Gene Transcription and Neuronal Morphology. J. Neurosci. Off. J. Soc. Neurosci. 2005, 25, 7821–7830. [Google Scholar] [CrossRef]
  232. Speranza, L.; Labus, J.; Volpicelli, F.; Guseva, D.; Lacivita, E.; Leopoldo, M.; Bellenchi, G.C.; Di Porzio, U.; Bijata, M.; Perrone-Capano, C.; et al. Serotonin 5- HT 7 Receptor Increases the Density of Dendritic Spines and Facilitates Synaptogenesis in Forebrain Neurons. J. Neurochem. 2017, 141, 647–661. [Google Scholar] [CrossRef] [PubMed]
  233. Sanger, G.J. 5-Hydroxytryptamine and the Gastrointestinal Tract: Where Next? Trends Pharmacol. Sci. 2008, 29, 465–471. [Google Scholar] [CrossRef] [PubMed]
  234. Ciranna, L.; Catania, M.V. 5-HT7 Receptors as Modulators of Neuronal Excitability, Synaptic Transmission and Plasticity: Physiological Role and Possible Implications in Autism Spectrum Disorders. Front. Cell. Neurosci. 2014, 8, 250. [Google Scholar] [CrossRef]
  235. Chang Chien, C.-C.; Hsin, L.-W.; Su, M.-J. Activation of Serotonin 5-HT7 Receptor Induces Coronary Flow Increase in Isolated Rat Heart. Eur. J. Pharmacol. 2015, 748, 68–75. [Google Scholar] [CrossRef]
  236. Hedlund, P.B.; Huitron-Resendiz, S.; Henriksen, S.J.; Sutcliffe, J.G. 5-HT7 Receptor Inhibition and Inactivation Induce Antidepressantlike Behavior and Sleep Pattern. Biol. Psychiatry 2005, 58, 831–837. [Google Scholar] [CrossRef]
  237. Blattner, K.M.; Canney, D.J.; Pippin, D.A.; Blass, B.E. Pharmacology and Therapeutic Potential of the 5-HT7 Receptor. ACS Chem. Neurosci. 2019, 10, 89–119. [Google Scholar] [CrossRef]
  238. Shimizu, M.; Nishida, A.; Zensho, H.; Miyata, M.; Yamawaki, S. Agonist-Induced Desensitization of Adenylyl Cyclase Activity Mediated by 5-Hydroxytryptamine7 Receptors in Rat Frontocortical Astrocytes. Brain Res. 1998, 784, 57–62. [Google Scholar] [CrossRef] [PubMed]
  239. Mahé, C.; Loetscher, E.; Dev, K.K.; Bobirnac, I.; Otten, U.; Schoeffter, P. Serotonin 5-HT7 Receptors Coupled to Induction of Interleukin-6 in Human Microglial MC-3 Cells. Neuropharmacology 2005, 49, 40–47. [Google Scholar] [CrossRef]
  240. Hashemi-Firouzi, N.; Komaki, A.; Soleimani Asl, S.; Shahidi, S. The Effects of the 5-HT7 Receptor on Hippocampal Long-Term Potentiation and Apoptosis in a Rat Model of Alzheimer’s Disease. Brain Res. Bull. 2017, 135, 85–91. [Google Scholar] [CrossRef]
  241. Wan, M.; Ding, L.; Wang, D.; Han, J.; Gao, P. Serotonin: A Potent Immune Cell Modulator in Autoimmune Diseases. Front. Immunol. 2020, 11, 186. [Google Scholar] [CrossRef]
  242. Leff-Gelman, P.; Mancilla-Herrera, I.; Flores-Ramos, M.; Cruz-Fuentes, C.; Reyes-Grajeda, J.P.; García-Cuétara, M.D.P.; Bugnot-Pérez, M.D.; Pulido-Ascencio, D.E. The Immune System and the Role of Inflammation in Perinatal Depression. Neurosci. Bull. 2016, 32, 398–420. [Google Scholar] [CrossRef] [PubMed]
  243. Abuelezz, S.A.; Hendawy, N.; Magdy, Y. Targeting Oxidative Stress, Cytokines and Serotonin Interactions Via Indoleamine 2, 3 Dioxygenase by Coenzyme Q10: Role in Suppressing Depressive Like Behavior in Rats. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2017, 12, 277–291. [Google Scholar] [CrossRef] [PubMed]
  244. Schneider-Schaulies, J.; Beyersdorf, N. CD4+ Foxp3+ Regulatory T Cell-Mediated Immunomodulation by Anti-Depressants Inhibiting Acid Sphingomyelinase. Biol. Chem. 2018, 399, 1175–1182. [Google Scholar] [CrossRef]
  245. Stasi, C.; Sadalla, S.; Milani, S. The Relationship Between the Serotonin Metabolism, Gut-Microbiota and the Gut-Brain Axis. Curr. Drug Metab. 2019, 20, 646–655. [Google Scholar] [CrossRef]
  246. Hersey, M.; Woodruff, J.L.; Maxwell, N.; Sadek, A.T.; Bykalo, M.K.; Bain, I.; Grillo, C.A.; Piroli, G.G.; Hashemi, P.; Reagan, L.P. High-Fat Diet Induces Neuroinflammation and Reduces the Serotonergic Response to Escitalopram in the Hippocampus of Obese Rats. Brain. Behav. Immun. 2021, 96, 63–72. [Google Scholar] [CrossRef] [PubMed]
  247. Wróbel, A.; Szklarczyk, J.; Barańska, I.; Majda, A.; Jaworek, J. Association between Levels of Serotonin, Melatonin, Cortisol and the Clinical Condition of Patients with Rheumatoid Arthritis. Rheumatol. Int. 2023, 43, 859–866. [Google Scholar] [CrossRef]
  248. Han, K.M.; Ham, B.J. How Inflammation Affects the Brain in Depression: A Review of Functional and Structural MRI Studies. J. Clin. Neurol. Seoul Korea 2021, 17, 503–515. [Google Scholar] [CrossRef]
  249. Haq, S.; Grondin, J.A.; Khan, W.I. Tryptophan-Derived Serotonin-Kynurenine Balance in Immune Activation and Intestinal Inflammation. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2021, 35, e21888. [Google Scholar] [CrossRef] [PubMed]
  250. Tchinda Defo, S.H.; Moussa, D.; Bouvourné, P.; Guédang Nyayi, S.D.; Woumitna, G.C.; Kodji, K.; Wado, E.K.; Ngatanko Abaissou, H.H.; Foyet, H.S. Unpredictable Chronic Mild Stress Induced Anxio-Depressive Disorders and Enterobacteria Dysbiosis: Potential Protective Effects of Detariummicrocarpum. J. Ethnopharmacol. 2025, 337, 118940. [Google Scholar] [CrossRef]
  251. Anderson, G. Depression Pathophysiology: Astrocyte Mitochondrial Melatonergic Pathway as Crucial Hub. Int. J. Mol. Sci. 2022, 24, 350. [Google Scholar] [CrossRef]
  252. Savonije, K.; Weaver, D.F. The Role of Tryptophan Metabolism in Alzheimer’s Disease. Brain Sci. 2023, 13, 292. [Google Scholar] [CrossRef]
  253. Sheline, Y.I.; West, T.; Yarasheski, K.; Swarm, R.; Jasielec, M.S.; Fisher, J.R.; Ficker, W.D.; Yan, P.; Xiong, C.; Frederiksen, C.; et al. An Antidepressant Decreases CSF Aβ Production in Healthy Individuals and in Transgenic AD Mice. Sci. Transl. Med. 2014, 6, 236re4. [Google Scholar] [CrossRef]
  254. Singh, K.; Gupta, J.K.; Sethi, P.; Mathew, S.; Bhatt, A.; Sharma, M.C.; Saha, S.; Shamim; Kumar, S. Recent Advances in the Synthesis of Antioxidant Derivatives: Pharmacological Insights for Neurological Disorders. Curr. Top. Med. Chem. 2024, 24, 1940–1959. [Google Scholar] [CrossRef] [PubMed]
  255. Darras, F.H.; Pockes, S.; Huang, G.; Wehle, S.; Strasser, A.; Wittmann, H.-J.; Nimczick, M.; Sotriffer, C.A.; Decker, M. Synthesis, Biological Evaluation, and Computational Studies of Tri- and Tetracyclic Nitrogen-Bridgehead Compounds as Potent Dual-Acting AChE Inhibitors and h H3 Receptor Antagonists. ACS Chem. Neurosci. 2014, 5, 225–242. [Google Scholar] [CrossRef] [PubMed]
  256. Dolles, D.; Nimczick, M.; Scheiner, M.; Ramler, J.; Stadtmüller, P.; Sawatzky, E.; Drakopoulos, A.; Sotriffer, C.; Wittmann, H.-J.; Strasser, A.; et al. Aminobenzimidazoles and Structural Isomers as Templates for Dual-Acting Butyrylcholinesterase Inhibitors and hCB2 R Ligands To Combat Neurodegenerative Disorders. ChemMedChem 2016, 11, 1270–1283. [Google Scholar] [CrossRef] [PubMed]
  257. Dolles, D.; Hoffmann, M.; Gunesch, S.; Marinelli, O.; Möller, J.; Santoni, G.; Chatonnet, A.; Lohse, M.J.; Wittmann, H.-J.; Strasser, A.; et al. Structure–Activity Relationships and Computational Investigations into the Development of Potent and Balanced Dual-Acting Butyrylcholinesterase Inhibitors and Human Cannabinoid Receptor 2 Ligands with Pro-Cognitive in Vivo Profiles. J. Med. Chem. 2018, 61, 1646–1663. [Google Scholar] [CrossRef] [PubMed]
  258. Szałaj, N.; Godyń, J.; Jończyk, J.; Pasieka, A.; Panek, D.; Wichur, T.; Więckowski, K.; Zaręba, P.; Bajda, M.; Pislar, A.; et al. Multidirectional in Vitro and in Cellulo Studies as a Tool for Identification of Multi-Target-Directed Ligands Aiming at Symptoms and Causes of Alzheimer’s Disease. J. Enzym. Inhib. Med. Chem. 2020, 35, 1944–1952. [Google Scholar] [CrossRef]
  259. Rochais, C.; Lecoutey, C.; Gaven, F.; Giannoni, P.; Hamidouche, K.; Hedou, D.; Dubost, E.; Genest, D.; Yahiaoui, S.; Freret, T.; et al. Novel Multitarget-Directed Ligands (MTDLs) with Acetylcholinesterase (AChE) Inhibitory and Serotonergic Subtype 4 Receptor (5-HT4R) Agonist Activities as Potential Agents against Alzheimer’s Disease: The Design of Donecopride. J. Med. Chem. 2015, 58, 3172–3187. [Google Scholar] [CrossRef]
  260. Dumuis, A.; Sebben, M.; Monferini, E.; Nicola, M.; Turconi, M.; Ladinsky, H.; Bockaert, J. Azabicycloalkyl Benzimidazolone Derivatives as a Novel Class of Potent Agonists at the 5-HT4 Receptor Positively Coupled to Adenylate Cyclase in Brain. Naunyn. Schmiedebergs Arch. Pharmacol. 1991, 343, 245–251. [Google Scholar] [CrossRef]
  261. Manzke, T.; Guenther, U.; Ponimaskin, E.G.; Haller, M.; Dutschmann, M.; Schwarzacher, S.; Richter, D.W. 5-HT4(a) Receptors Avert Opioid-Induced Breathing Depression without Loss of Analgesia. Science 2003, 301, 226–229. [Google Scholar] [CrossRef]
  262. Chelusnova, Y.V.; Voronina, P.A.; Belinskaia, D.A.; Goncharov, N.V. Benzimidazole-Carboxamides as Potential Therapeutics for Alzheimer’s Disease: Primary Analysis In Silico and In Vitro. Bull. Exp. Biol. Med. 2023, 175, 345–352. [Google Scholar] [CrossRef] [PubMed]
  263. Belinskaia, D.A.; Voronina, P.A.; Krivorotov, D.V.; Jenkins, R.O.; Goncharov, N.V. Anticholinesterase and Serotoninergic Evaluation of Benzimidazole-Carboxamides as Potential Multifunctional Agents for the Treatment of Alzheimer’s Disease. Pharmaceutics 2023, 15, 2159. [Google Scholar] [CrossRef] [PubMed]
  264. Choe, Y.M.; Suh, G.-H.; Lee, B.C.; Choi, I.-G.; Kim, H.S.; Kim, J.W.; Hwang, J.; Yi, D.; Kim, J.W. High-Intensity Walking in Midlife Is Associated with Improved Memory in Physically Capable Older Adults. Alzheimers Res. Ther. 2023, 15, 143. [Google Scholar] [CrossRef]
  265. Park, S.-S.; Park, H.-S.; Kim, C.-J.; Baek, S.-S.; Kim, T.-W. Exercise Attenuates Maternal Separation-Induced Mood Disorder-like Behaviors by Enhancing Mitochondrial Functions and Neuroplasticity in the Dorsal Raphe. Behav. Brain Res. 2019, 372, 112049. [Google Scholar] [CrossRef] [PubMed]
  266. Sampaio, A.; Marques, E.A.; Mota, J.; Carvalho, J. Effects of a Multicomponent Exercise Program in Institutionalized Elders with Alzheimer’s Disease. Dementia 2019, 18, 417–431. [Google Scholar] [CrossRef] [PubMed]
  267. Liu, Y.; Chu, J.M.T.; Yan, T.; Zhang, Y.; Chen, Y.; Chang, R.C.C.; Wong, G.T.C. Short-Term Resistance Exercise Inhibits Neuroinflammation and Attenuates Neuropathological Changes in 3xTg Alzheimer’s Disease Mice. J. Neuroinflamm. 2020, 17, 4. [Google Scholar] [CrossRef]
  268. Lv, S.; Wang, Q.; Liu, W.; Zhang, X.; Cui, M.; Li, X.; Xu, Y. Comparison of Various Exercise Interventions on Cognitive Function in Alzheimer’s Patients: A Network Meta-Analysis. Arch. Gerontol. Geriatr. 2023, 115, 105113. [Google Scholar] [CrossRef]
  269. Adan, R.A.H.; van der Beek, E.M.; Buitelaar, J.K.; Cryan, J.F.; Hebebrand, J.; Higgs, S.; Schellekens, H.; Dickson, S.L. Nutritional Psychiatry: Towards Improving Mental Health by What You Eat. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2019, 29, 1321–1332. [Google Scholar] [CrossRef]
  270. Goncharov, N.V.; Belinskaia, D.A.; Ukolov, A.I.; Jenkins, R.O.; Avdonin, P.V. Organosulfur compounds as nutraceuticals. In Nutraceuticals: Efficacy, Safety, and Toxicity, 2nd ed.; Gupta, R.C., Lall, R., Srivastava, A., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 911–924. ISBN 978-0-12-821038-3. [Google Scholar] [CrossRef]
  271. Tarozzi, A.; Angeloni, C.; Malaguti, M.; Morroni, F.; Hrelia, S.; Hrelia, P. Sulforaphane as a Potential Protective Phytochemical against Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2013, 2013, 415078. [Google Scholar] [CrossRef]
  272. Colín-González, A.L.; Santana, R.A.; Silva-Islas, C.A.; Chánez-Cárdenas, M.E.; Santamaría, A.; Maldonado, P.D. The Antioxidant Mechanisms Underlying the Aged Garlic Extract- and S-Allylcysteine-Induced Protection. Oxid. Med. Cell. Longev. 2012, 2012, 907162. [Google Scholar] [CrossRef]
  273. Wang, L.; Jiang, L.; Chu, Y.; Feng, F.; Tang, W.; Chen, C.; Qiu, Y.; Hu, Z.; Diao, H.; Tang, Z. Dietary Taurine Improves Growth Performance and Intestine Health via the GSH/GSSG Antioxidant System and Nrf2/ARE Signaling Pathway in Weaned Piglets. Antioxidants 2023, 12, 1852. [Google Scholar] [CrossRef] [PubMed]
  274. Seol, S.-I.; Kang, I.S.; Lee, J.S.; Lee, J.-K.; Kim, C. Taurine Chloramine-Mediated Nrf2 Activation and HO-1 Induction Confer Protective Effects in Astrocytes. Antioxidants 2024, 13, 169. [Google Scholar] [CrossRef] [PubMed]
  275. Bellavite, P. Neuroprotective Potentials of Flavonoids: Experimental Studies and Mechanisms of Action. Antioxidants 2023, 12, 280. [Google Scholar] [CrossRef] [PubMed]
  276. Zhang, J.; Zhou, X.; Wu, W.; Wang, J.; Xie, H.; Wu, Z. Regeneration of Glutathione by α-Lipoic Acid via Nrf2/ARE Signaling Pathway Alleviates Cadmium-Induced HepG2 Cell Toxicity. Environ. Toxicol. Pharmacol. 2017, 51, 30–37. [Google Scholar] [CrossRef]
  277. Toohey, J.I.; Cooper, A.J.L. Thiosulfoxide (Sulfane) Sulfur: New Chemistry and New Regulatory Roles in Biology. Molecules 2014, 19, 12789–12813. [Google Scholar] [CrossRef]
  278. Satoh, T.; McKercher, S.R.; Lipton, S.A. Reprint of: Nrf2/ARE-Mediated Antioxidant Actions of pro-Electrophilic Drugs. Free Radic. Biol. Med. 2014, 66, 45–57. [Google Scholar] [CrossRef]
Figure 1. The barrel-like structure of AβO forms ion channels in the cell membrane, allowing Ca2+ to enter the cell. Ca2+ binds to VDAC, regulating its permeability and inhibiting ATP release into the cytosol. Aβ/AβO  enter the mitochondrial matrix (via TOM and TIM translocators localized on the outer and inner membranes) and interact directly with complex I of the respiratory chain, disrupting ETC function, decreasing ATP, and increasing ROS. The decrease in ATP leads to an increase in the (NAD+/NADH) ratio and activation of SIRT1. ROS production and the activation of JNK kinase are increased. SIRT1 and JNK penetrate the nucleus and activate PGC-1α. Serotonin regulation of mitochondrial biogenesis and function may be mediated by the 5-HT2A receptor and PGC-1α, the master modulator of mitochondrial biogenesis. Activation of the 5-HT2A receptor on neurons is accompanied by the stimulation of PGC-1α through several signaling pathways: (1) αq—PLCβ—IP3—IP3R—↑Ca2+—↑CaM—↑CN—NFAT; (2) PLCβ—DAG—PKC—JNK; (3) GRK—β-arrestin—β-arrestin/Src—MAPK—ERK1/2; (4) αq—PLCβ—IP3—IP3R—↑Ca2+—CaM—↑CaMK—CREB. Abbreviations: Aβ—amyloid-β; AβO—Aβ oligomer; αq—subunit of heterotrimer protein Gq; ATP—adenosine triphosphate; CaM—calmodulin; CaMK—calmodulin kinase; CN—calcineurin; CREB—cAMP-response element-binding protein; DAG—diacylglycerol; ERRa—estrogen-related receptor alpha; ERK—extracellular signal regulated kinase; ETC—electron transport chain; GRK—G-protein-coupled receptor kinases; 5-HT2A—serotonin G-protein-coupled receptor, subtype 2A; IP3—inositol trisphosphate; IP3R—receptor of inositol trisphosphate; mtDNA—mitochondrial DNA; JNK—Jun amino-terminal kinase; MAPK—mitogen-activated protein kinase; MBioG—mitochondrial biogenesis; NFAT—nuclear factor of activated T cells; NRF1/2—nuclear respiratory factors 1/2; PGC-1α—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PLCβ—phospholipase C β; PKC—protein kinase C; ROS—reactive oxygen species; SIRT1—NAD+-dependent deacetylase sirtuin-1; Src—non-receptor tyrosine kinase; TIM—inner-membrane translocase; TOM—outer-membrane translocase; TFAM—transcription factor A, mitochondrial; VDAC—voltage-dependent anion channel.
Figure 1. The barrel-like structure of AβO forms ion channels in the cell membrane, allowing Ca2+ to enter the cell. Ca2+ binds to VDAC, regulating its permeability and inhibiting ATP release into the cytosol. Aβ/AβO  enter the mitochondrial matrix (via TOM and TIM translocators localized on the outer and inner membranes) and interact directly with complex I of the respiratory chain, disrupting ETC function, decreasing ATP, and increasing ROS. The decrease in ATP leads to an increase in the (NAD+/NADH) ratio and activation of SIRT1. ROS production and the activation of JNK kinase are increased. SIRT1 and JNK penetrate the nucleus and activate PGC-1α. Serotonin regulation of mitochondrial biogenesis and function may be mediated by the 5-HT2A receptor and PGC-1α, the master modulator of mitochondrial biogenesis. Activation of the 5-HT2A receptor on neurons is accompanied by the stimulation of PGC-1α through several signaling pathways: (1) αq—PLCβ—IP3—IP3R—↑Ca2+—↑CaM—↑CN—NFAT; (2) PLCβ—DAG—PKC—JNK; (3) GRK—β-arrestin—β-arrestin/Src—MAPK—ERK1/2; (4) αq—PLCβ—IP3—IP3R—↑Ca2+—CaM—↑CaMK—CREB. Abbreviations: Aβ—amyloid-β; AβO—Aβ oligomer; αq—subunit of heterotrimer protein Gq; ATP—adenosine triphosphate; CaM—calmodulin; CaMK—calmodulin kinase; CN—calcineurin; CREB—cAMP-response element-binding protein; DAG—diacylglycerol; ERRa—estrogen-related receptor alpha; ERK—extracellular signal regulated kinase; ETC—electron transport chain; GRK—G-protein-coupled receptor kinases; 5-HT2A—serotonin G-protein-coupled receptor, subtype 2A; IP3—inositol trisphosphate; IP3R—receptor of inositol trisphosphate; mtDNA—mitochondrial DNA; JNK—Jun amino-terminal kinase; MAPK—mitogen-activated protein kinase; MBioG—mitochondrial biogenesis; NFAT—nuclear factor of activated T cells; NRF1/2—nuclear respiratory factors 1/2; PGC-1α—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PLCβ—phospholipase C β; PKC—protein kinase C; ROS—reactive oxygen species; SIRT1—NAD+-dependent deacetylase sirtuin-1; Src—non-receptor tyrosine kinase; TIM—inner-membrane translocase; TOM—outer-membrane translocase; TFAM—transcription factor A, mitochondrial; VDAC—voltage-dependent anion channel.
Ijms 26 05218 g001
Figure 2. Serotonin and BDNF initiate neurogenesis. Adult hippocampal neurogenesis (AHN) persists throughout life in mammals, including humans. AHN is the creation (from neural stem cells (NSCs)) of new neurons, which integrate into the existing neural circuit of the adult brain. Most NSCs are concentrated in the dentate gyrus of the hippocampus and the subventricular zone (SVZ). The dentate gyrus of the hippocampus is one of the brain regions affected first in patients with Alzheimer’s disease. Neurogenic processes, including cell proliferation, differentiation, migration and maturation, are led by neurotrophic factors. Expression of BDNF in serotoninergic neurons increases stress tolerance and stimulates hippocampal neurogenesis in adults. BDNF activates the TrKB receptor, which acts via protein kinase C to activate proteins involved in cell survival and cell migration. The hippocampus expresses various 5-HT receptor subtypes, most of which (i.e., 5-HT1A, 2A, 3, 4, and 6) are expressed in the DG subregion of the hippocampus. The 5-HT2A receptor is the main subtype of serotonin receptors mediating its excitatory action. 5-HT2A receptors are localized predominantly in the plasma membrane and also enter the cell under the action of beta-arrestin. This results in the activation of NF-kB and NFAT, and mitochondrial biogenesis. The 5-HT1A receptor activates the CREB and K+ channel. Selective stimulation of 5-HT1A receptors increases the number of proliferating cells. Activation of the 5-HT3 receptor leads to membrane depolarization, the opening of voltage-dependent Ca2+ channels, and activation of calcineurin phosphatase and the NFAT transcription factor. 5-HT4 initiates the expression of BDNF. The 5-HT6 receptor is involved in the initiation of neurite outgrowth (NeuOG) by the agonist independently activating the cyclin dependent kinase 5 (Cdk5)-Cdc42 signaling pathway. The 5-HT signaling pathway activates the CREB, which promotes the transcription of the BDNF gene. Abbreviations: AC—adenylate cyclase; αq—subunit of heterotrimer protein Gq; BDNF—brain-derived neurotrophic factor; CaM—calmodulin; cAMP—cyclic adenosine monophosphate; CaMK—calmodulin kinase; CN—calcineurin; Cdk5—cyclin-dependent kinase; CREB—cAMP-response element-binding protein; DAG—diacylglycerol; DPM—plasma membrane depolarization; ERK—extracellular signal regulated kinase; GIRK—G-protein-coupled inwardly rectifying potassium channels; GRB2—adaptor proteins containing SH2 and SH3 domains; G—heterotrimer protein Gi; 5-HT—5-hydroxytryptamine (serotonin); IP3—inositol trisphosphate; IP3R—receptor of inositol trisphosphate; L-type CC—L-type calcium channel; MEK1/2—dual-specificity kinase; MERK—protein kinase; NeuOGs—neurite outgrows; NFAT—nuclear factor of activated T cells; NF-kB—nuclear factor kappa-light-chain-enhancer of activated B cells; p35—universal caspase inhibitor; PDK1 or 2—phospholipid-dependent kinase 1 or 2; PIP2—phosphatidylinositol diphosphate; PI3K—phosphatidylinositol 3 kinase; PLCγ—phospholipase Cγ; PKA—protein kinase A; PKB—protein kinase B; PKC—protein kinase C; PM—phosphatase; Raf—serine/threonine kinase; Ras—small G-protein, GTP-ase; RSK—ribosomal S6 kinase; SHC—Src homology 2 domain containing transforming proteins; SOS—guanine nucleotide exchange factor; TrKB—tropomyosin receptor kinase B; VMAT2—vesicular monoamine transporter 2.
Figure 2. Serotonin and BDNF initiate neurogenesis. Adult hippocampal neurogenesis (AHN) persists throughout life in mammals, including humans. AHN is the creation (from neural stem cells (NSCs)) of new neurons, which integrate into the existing neural circuit of the adult brain. Most NSCs are concentrated in the dentate gyrus of the hippocampus and the subventricular zone (SVZ). The dentate gyrus of the hippocampus is one of the brain regions affected first in patients with Alzheimer’s disease. Neurogenic processes, including cell proliferation, differentiation, migration and maturation, are led by neurotrophic factors. Expression of BDNF in serotoninergic neurons increases stress tolerance and stimulates hippocampal neurogenesis in adults. BDNF activates the TrKB receptor, which acts via protein kinase C to activate proteins involved in cell survival and cell migration. The hippocampus expresses various 5-HT receptor subtypes, most of which (i.e., 5-HT1A, 2A, 3, 4, and 6) are expressed in the DG subregion of the hippocampus. The 5-HT2A receptor is the main subtype of serotonin receptors mediating its excitatory action. 5-HT2A receptors are localized predominantly in the plasma membrane and also enter the cell under the action of beta-arrestin. This results in the activation of NF-kB and NFAT, and mitochondrial biogenesis. The 5-HT1A receptor activates the CREB and K+ channel. Selective stimulation of 5-HT1A receptors increases the number of proliferating cells. Activation of the 5-HT3 receptor leads to membrane depolarization, the opening of voltage-dependent Ca2+ channels, and activation of calcineurin phosphatase and the NFAT transcription factor. 5-HT4 initiates the expression of BDNF. The 5-HT6 receptor is involved in the initiation of neurite outgrowth (NeuOG) by the agonist independently activating the cyclin dependent kinase 5 (Cdk5)-Cdc42 signaling pathway. The 5-HT signaling pathway activates the CREB, which promotes the transcription of the BDNF gene. Abbreviations: AC—adenylate cyclase; αq—subunit of heterotrimer protein Gq; BDNF—brain-derived neurotrophic factor; CaM—calmodulin; cAMP—cyclic adenosine monophosphate; CaMK—calmodulin kinase; CN—calcineurin; Cdk5—cyclin-dependent kinase; CREB—cAMP-response element-binding protein; DAG—diacylglycerol; DPM—plasma membrane depolarization; ERK—extracellular signal regulated kinase; GIRK—G-protein-coupled inwardly rectifying potassium channels; GRB2—adaptor proteins containing SH2 and SH3 domains; G—heterotrimer protein Gi; 5-HT—5-hydroxytryptamine (serotonin); IP3—inositol trisphosphate; IP3R—receptor of inositol trisphosphate; L-type CC—L-type calcium channel; MEK1/2—dual-specificity kinase; MERK—protein kinase; NeuOGs—neurite outgrows; NFAT—nuclear factor of activated T cells; NF-kB—nuclear factor kappa-light-chain-enhancer of activated B cells; p35—universal caspase inhibitor; PDK1 or 2—phospholipid-dependent kinase 1 or 2; PIP2—phosphatidylinositol diphosphate; PI3K—phosphatidylinositol 3 kinase; PLCγ—phospholipase Cγ; PKA—protein kinase A; PKB—protein kinase B; PKC—protein kinase C; PM—phosphatase; Raf—serine/threonine kinase; Ras—small G-protein, GTP-ase; RSK—ribosomal S6 kinase; SHC—Src homology 2 domain containing transforming proteins; SOS—guanine nucleotide exchange factor; TrKB—tropomyosin receptor kinase B; VMAT2—vesicular monoamine transporter 2.
Ijms 26 05218 g002
Figure 3. Relationships between serotonin, depression, neurodegeneration, AD, and the circadian clock system. 5-HT regulates circadian rhythms by acting via 5-HT1B and 5-HT7 receptors. Disruption of circadian rhythms can lead to sleep disturbances, depression, decreased BDNF, mitochondrial dysfunction, increased ROS, and oxidative stress. Depression impairs glymphatic system activity, promotes AβO formation and accumulation, and initiates AD. Abbreviations: Aβ—amyloid-β; AβO—β-amyloid oligomer; AD—Alzheimer’s disease; 5-HT—serotonin; BDNF—brain-derived neurotrophic factor; ROS—reactive oxygen species; NE—norepinephrine.
Figure 3. Relationships between serotonin, depression, neurodegeneration, AD, and the circadian clock system. 5-HT regulates circadian rhythms by acting via 5-HT1B and 5-HT7 receptors. Disruption of circadian rhythms can lead to sleep disturbances, depression, decreased BDNF, mitochondrial dysfunction, increased ROS, and oxidative stress. Depression impairs glymphatic system activity, promotes AβO formation and accumulation, and initiates AD. Abbreviations: Aβ—amyloid-β; AβO—β-amyloid oligomer; AD—Alzheimer’s disease; 5-HT—serotonin; BDNF—brain-derived neurotrophic factor; ROS—reactive oxygen species; NE—norepinephrine.
Ijms 26 05218 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dolgacheva, L.P.; Zinchenko, V.P.; Nadeev, A.D.; Goncharov, N.V. Serotonergic Regulation in Alzheimer’s Disease. Int. J. Mol. Sci. 2025, 26, 5218. https://doi.org/10.3390/ijms26115218

AMA Style

Dolgacheva LP, Zinchenko VP, Nadeev AD, Goncharov NV. Serotonergic Regulation in Alzheimer’s Disease. International Journal of Molecular Sciences. 2025; 26(11):5218. https://doi.org/10.3390/ijms26115218

Chicago/Turabian Style

Dolgacheva, Lyudmila P., Valery P. Zinchenko, Alexander D. Nadeev, and Nikolay V. Goncharov. 2025. "Serotonergic Regulation in Alzheimer’s Disease" International Journal of Molecular Sciences 26, no. 11: 5218. https://doi.org/10.3390/ijms26115218

APA Style

Dolgacheva, L. P., Zinchenko, V. P., Nadeev, A. D., & Goncharov, N. V. (2025). Serotonergic Regulation in Alzheimer’s Disease. International Journal of Molecular Sciences, 26(11), 5218. https://doi.org/10.3390/ijms26115218

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

Article Metrics

Back to TopTop