Next Article in Journal
Impact of Endocrine Disruptors upon Non-Genetic Inheritance
Previous Article in Journal
Pathophysiology of Atherosclerosis
Previous Article in Special Issue
New Insights into Multiple Sclerosis Mechanisms: Lipids on the Track to Control Inflammation and Neurodegeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Structural Plasticity of the Hippocampus in Neurodegenerative Diseases

by
Poornima D. E. Weerasinghe-Mudiyanselage
1,†,
Mary Jasmin Ang
1,2,†,
Sohi Kang
1,
Joong-Sun Kim
1 and
Changjong Moon
1,*
1
Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea
2
College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(6), 3349; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23063349
Submission received: 25 February 2022 / Revised: 17 March 2022 / Accepted: 18 March 2022 / Published: 20 March 2022
(This article belongs to the Special Issue New Mechanisms and Therapeutics in Neurological Diseases)

Abstract

:
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.

1. Introduction

Neuroplasticity is the ability of neural networks in the brain to alter through development and rearrangement, and is generally categorized into structural and functional plasticity [1,2,3]. Structural plasticity involves the expansion or retraction of the synaptic area through the remodeling of spines, dendrites, and/or axons [1]. Functional plasticity involves regulation of neurotransmission, reorganization of synaptic components and receptors, and regulation of the strength or efficiency of synaptic transmission [4,5].
Structural plasticity, such as dendritic formation and spine development, is mainly exhibited through the regulation of the neuronal actin cytoskeleton [6,7]. Several actin regulating proteins influence the structural plasticity of neurons by maintaining the equilibrium between G- and F-actin [8]. Furthermore, several signaling cascades are involved in neuronal structural modification, wherein actin and microtubule cytoskeletons serve as the common end targets [9,10]. Guanosine triphosphate hydroxylase (GTPases) is an important family of signaling molecules that transduces extracellular signals to control the actin assembly [11]. Ras-related C3 botulinum toxin substrate 1 (Rac1) and Ras homolog (Rho) family member A (RhoA) are some widely studied members of the Rho family of GTPases. Rac1 promotes spine growth through the activation of p21 (RAC1) activated kinase 1 (PAK1) and LIM domain kinase 1 (LIMK1), while RhoA negatively regulates the spine formation [7]. Two major upstream signals which can trigger GTPases are the brain-derived neurotrophic factors (BDNF) [12,13] and glutamate receptors (GluRs) [14], both of which greatly affect the neuronal micromorphometry [12,14,15]. For example, BDNF promotes the actin polymerization in dendritic spines of hippocampal neurons through Rac1 [13]. The stimulation of group I metabotropic GluRs in hippocampal neurons can lead to beneficial effects on neuroplasticity [14], while excessive activation of ionotropic GluRs results in deleterious effects [16,17]. In addition, spine morphology is also regulated by the same synaptic proteins that regulate actin and microtubules in the dendrite [18]. For example, the post synaptic density (PSD) is enlarged due to an increase in PSD protein95 (PSD95), which facilitates an early structural enlargement of dendritic spines [18,19]. BDNF-induced increase in PSD95 leads to microtubule rearrangements during the maturation period of dendritic spines [19]. Additionally, an increase in polysialylated-neural cell adhesion molecules promotes synaptic remodeling during persistent types of plasticity [20].
Functional plasticity is commonly interchanged with “synaptic plasticity” due to its primary involvement in synaptic transmission [21,22,23,24]. Synaptic plasticity may operate in the following ways: (1) growth of new synaptic connections or pruning of existing ones, (2) modification of the strength or efficacy of synaptic transmission, and (3) modulation of the excitability of existing synapses. Due to the diversity of neuronal functions, many forms of synaptic plasticity have been described, such as post-tetanic potentiation (PTP), long-term potentiation (LTP), and long-term depression (LTD) [24]. During synaptic potentiation, receptors are translocated to the post-synaptic membrane where they interact with other members of the PSD; however, during synaptic depression, the receptors are endocytosed [20]. Various signaling pathways (such as mitogen-activated protein kinases [MAPK] pathway, Wnt pathway, and BDNF-TrkB signaling) influence the induction of activity-dependent synaptic plasticity [25,26,27]. Such activity-dependent synaptic plasticity has been studied in experimental setups, where synaptic plasticity was induced through high- or low-frequency stimulation of neuronal afferents that cause LTP [28,29] and LTD [30] during synaptic transmission. The interplay of structural and functional plasticity is evidenced by synaptic plasticity that leads to structural modifications in dendritic spines; induction of LTP that promotes spine head growth, formation, and maintenance. However, LTD causes the spine heads to shrink and retract [31]. This relationship between structural and functional plasticity underlies vital processes that maintain the normal anatomy and physiology of the brain.
Volumetric and morphological changes in dendritic structures are widely studied in various physiological and pathological conditions [32,33]. Regulation of neuroplasticity in dendrites, spines, and synapses are sensitive to both positive (learning [34] and environmental enrichment [35]) and negative stimuli (radiation [36,37], stress [38], chemical exposure [39], aging [40,41,42] and neurodegenerative disease [43,44]). Moreover, neurodegenerative diseases, in particular, are described by dysregulations in the structural plasticity [45,46]. Dendritic and spine changes caused due to acute or chronic disturbances in brain tissue homeostasis may form the basis of neurodegenerative diseases [47]. The involvement of neuroplasticity in neurodegenerative diseases has been widely studied [45,48,49,50], as these changes in dendritic and spine structure in several brain regions serve as mechanisms for motor and non-motor features of various neurodegenerative diseases [51,52,53,54]. Additionally, aging is considered as a primary risk factor for the development of neurodegenerative diseases [55]; in fact, several review articles have already described dendritic structural alterations in the context of aging [56,57,58].
The hippocampus is an important brain region for its impressive capacity for lifelong plasticity [59]. Structural plasticity in the hippocampus is susceptible to a wide range of stimuli, many of which appear to have long-term impacts. For example, the rapid formation of new spines and enlargement of existing spines in hippocampal slices are associated with LTP-mediated increase in synaptic strength [9], whereas loss of post-synaptic actin and spine shrinkage are associated with LTD-induced actin depolymerization [60]. Furthermore, hippocampal neurons readily respond to environmental cues by showing differential pattern changes in dendritic and spine morphology [61]. The hippocampus is one of the brain areas that shows neuronal remodeling under several neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), multiple sclerosis (MS), vascular dementia (VD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS) [62,63,64,65,66,67].
AD, PD, and HD are highly prevalent neurodegenerative diseases [68,69], while MS is a non-traumatic, neuroinflammatory, and neurodegenerative disease affecting young adults [70]. VD and FTD are forms of dementia, which is a degenerative disease primarily characterized by intellectual and cognitive impairment [66]. ALS is a neurodegenerative disease with varying degrees of impairment of motor function and cognitive profile [71,72]. To date, numerous therapeutic approaches have been developed to relieve clinical symptoms in these diseases. However, non-motor dysfunctions respond poorly to conventional therapeutics and affect the quality of life (QOL) of the patients [73,74,75]. As a result, there is a need for improvement in the existing treatments or the development of entirely new strategies. The hippocampus has been the focus of neuroplasticity research for common neurodegenerative diseases, including AD, PD, HD, MS, VD, FTD, and ALS since it is a principal brain region for cognitive and emotional functions [76,77,78,79,80]. Several reviews [81,82,83] have already explored the involvement of functional or synaptic plasticity of the hippocampus in neurodegenerative diseases. Thus, in this review, we primarily focused on the role of structural plasticity of the hippocampus in neurodegenerative diseases.
In this study, articles describing structural plasticity of the hippocampus in neurodegenerative diseases, including AD, PD, HD, MS, VD, FTD, and ALS, were collected through a literature search using Google Scholar, PubMed, Web of Science, and Scopus. This review provides an overview of how structural plasticity in the hippocampus is involved in clinical and preclinical studies of neurodegenerative diseases. Further, this review suggests possible therapeutic approaches to regulate altered hippocampal neuroplasticity in neurodegenerative diseases. Additionally, the prevailing gaps in the knowledge of hippocampal neuroplasticity in neurodegenerative diseases will also be revealed to serve as starting points for future studies.

2. Hippocampal Dysfunction in Neurodegenerative Diseases

2.1. Alzheimer’s Disease

AD is one of the most prevalent neurodegenerative diseases with major neuropathological hallmarks as follows: (1) intra-neuronal accumulation of neurofibrillary tangles (NFTs) as a consequence of abnormal hyperphosphorylation of Tau (HPtau) proteins, (2) extracellular deposition of amyloid-β (Aβ) protein as senile plaques, and (3) massive neuronal death [84]. AD is an age-related disease featuring progressive loss of memory and cognitive abilities [85]. Individuals with AD usually undergo difficulties in learning, performance speed, recall accuracy, and problem-solving [86]. Hippocampal involvement in AD pathogenesis is evident from a wide range of clinical and preclinical studies [82,87,88,89,90,91]. In AD, the hippocampal cornu ammonis (CA) 1 subregion is the first target of pathological hallmarks (i.e., NFTs), followed by the subiculum, CA2, CA3, and dentate gyrus (DG) [92,93,94]. Particularly, overall neuronal loss in the CA1 region was predominantly observed in most of the studies that included patients with mild to severe AD [95,96,97]. Furthermore, impaired functional connectivity of the hippocampus with several other brain regions results in a combination of neuropsychological abnormalities in AD [80].
Cognitive impairment in early AD is largely due to synaptic dysfunction [98] (Figure 1). Causes of synaptic dysfunction in AD include decrease in overall cell count [99], synaptic deterioration due to soluble Aβ [100], synaptic pruning due to glial involvement [101], and significant decrease in synaptic vesicles [102,103]. Synaptic failure in the hippocampus is established in patients with AD as well as animal models [98,104]. In fact, hippocampal synaptic dysfunction is an early pathological hallmark in AD and is the underlying reason for memory impairment [105]. Hippocampal synaptic dysfunction associated with AD has been reported as decreased synaptic number [106,107,108], impaired synapse regeneration/synaptogenesis [109,110], loss of synaptic proteins (i.e., synaptophysin, synaptogyrin, synaptotagmin, syntaxin 1, PSD95, and Homer-1) [103,109,111,112,113,114,115], impaired LTP [108,111,113,114,116,117], and impaired synaptic signaling cascades (i.e., MAPK signaling and Wnt signaling) [118,119]. Memory impairment begins with subtle changes in hippocampus synaptic efficacy, before progressing to severe neuronal loss [120]. Interestingly, the synaptic dysfunction appears before the Aβ plaque formation, implying that a physiological deficit underlies the initial development of the disease [121,122,123].

2.2. Parkinson’s Disease

PD is the second most prevalent neurodegenerative disease in the world [124,125,126]. PD results from the deterioration of dopaminergic (DA) neurons in the extrapyramidal tract of the midbrain, along with the accumulation of alpha-synuclein (α-Syn) proteins (namely, Lewy bodies) in the nervous system. PD is a neurodegenerative condition known for both motor and non-motor symptoms [127]. Since the hallmark of PD is DA injury, much attention has been given to the structural, molecular, and functional changes in the nigrostriatal system. However, non-motor symptoms of PD that are associated with the hippocampus also significantly affect the QOL of the patients [128]. These symptoms include PD dementia, cognitive deficits, and sleep-wake disorders, which are evident in both the early phase and advanced stage of the PD [129,130]. Other non-motor symptoms including hyposmia, rapid eye movement, sleep behavior disorder, constipation, sadness, anxiety, depression, and apathy, may also occur before the beginning of motor deficits [131,132]. Several researchers suggest a key interaction between DA transmission and hippocampal neuroplasticity in non-motor symptoms; however, the complicated mechanisms underlying these symptoms may also involve non-DA systems. For example, hippocampal serotonergic [133,134] and noradrenergic (NA) [135] dysfunctions are reported in emotional dysregulation of PD.
Synaptic mechanisms underlying cognitive impairment in PD are widely reported in preclinical studies (Figure 1). For instance, impaired hippocampal synaptic plasticity in terms of decreased LTP is reported in neurotoxic models of PD, such as 6-hydroxydopamine (6-OHDA)-lesioned rats [136,137,138] and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mice [136,137,139]. Similarly, α-Syn transgenic (TG) mice also showed reduced hippocampal CA1 LTP [136]. Hippocampal neurons exhibited reduced excitatory post-synaptic current (EPSC) [140] and progressive impairments in neuronal excitability [100] upon α-Syn preformed fibrils (PFF) treatment. Synaptic protein alterations in CA1 [136,137] and DG regions [138] are also reported in 6-OHDA-lesioned rats and PFF-treated primary cultured hippocampal neurons [100]. Several studies suggest the importance of DA in the maintenance of hippocampal LTP through regulation of dopamine D1/5 receptors [141,142,143] and stimulation of synaptic protein synthesis [144,145], both of which demonstrate an interplay between the DA input and hippocampal synaptic plasticity for the maintenance of hippocampus-related functions. It has been reported that L-DOPA, also known as levodopa and l-3,4-dihydroxyphenylalanine, restored hippocampal LTP and alleviated cognitive impairments in clinical [146] and preclinical [129,136] conditions of PD. Although aberrations in functional plasticity are often attributed to DA signaling, other neurotransmitters such as NA [138,147] and serotonin [148] have also been implicated. Recent advances in studies exploring the genetic basis of PD-highlighted genes that may be related to the disruptions in synaptic vesicle endocytosis, and act as significant contributors to its pathogenesis. These endocytosis disruption-related genes include auxilin (DNAJC6) synaptojanin 1 (SYNJ1), endophilin A1 (SH3GL2), leucine-rich repeat kinase 2 (LRRK2), parkin (PRKN), and vacuolar protein sorting ortholog 35 (VPS35) [44].

2.3. Huntington’s Disease

HD is a progressive autosomal dominant neurodegenerative disorder, which is characterized by a combination of progressive motor and cognitive impairments, as well as neuropsychiatric symptoms [149,150,151]. These symptoms include distinct behavioral changes, including chorea, dystonia, cognitive decline, and emotional impairments [137,152]. HD results from expanded three nucleotide (Cytosine, Adenine, and Guanine)-repeats in the huntingtin gene [153]. The neuropathological changes in HD include atrophy and cell loss in the caudate putamen of the striatum, where medium spiny neurons are the most vulnerable [154,155,156]. This has led to the striatum being the center of attention for molecular, structural, and functional studies of HD [157,158,159]. However, other brain regions such as the hippocampus are also affected in HD [160]. Non-motor symptoms in preclinical and clinical HD conditions are significantly related to the hippocampus [78,161]. Mild cognitive impairments have been detected in patients with HD long before (up to 15 years) the formal diagnosis based on the motor symptoms [162]. Additionally, deficits in episodic memory, processing speed, executive function, and visuospatial perception were found in the clinical stages of HD [68]. Studies using environmental and pharmacological interventions imply that basal serotonergic disruption in the hippocampus is a causal factor in depressive-like behavior in HD [78]. Moreover, altered extracellular glutamate (Glu) dynamics in the hippocampus may also underpin cognitive deficits in HD [163].
Different forms of functional plasticity are impaired in human patients and mouse models of HD [164,165,166,167,168,169,170,171,172,173,174]. These abnormalities in synaptic function include both short- and long-term plasticity (Figure 1). For instance, impairments in long-term plasticity in terms of deficient induction and maintenance of LTP, as well as impairments in short-term plasticity in terms of decreased PTP [164,175,176] are reported in HD. Disruption of LTP at the hippocampal CA3-CA1 synapse is reported in Hu97/18-TG [171], proline-rich tyrosine kinase 2 (Pyk2)-deficient [172], and Q175FDN-knock-in mice [176]. LTP mediated by the N-methyl-D-aspartate receptors (NMDARs) at CA1 synapse in the hippocampus is also impaired in Pyk2-deficient [172], R6/2-TG [165], and R6/1-TG mice [177] at the symptomatic stage. These impairments are known to underlie cognitive disabilities, such as learning and memory [24,178,179]. Long-term deficits in the hippocampus are likely to be mediated by disturbances in a range of signaling pathways, contributing to the hippocampal dysfunction in HD [176]. Other possible factors mediating synaptic plasticity in HD are neurotransmitters such as DA, acetylcholine (Ach), and Glu [180], and neurotrophic factors such as BDNF [181].

2.4. Multiple Sclerosis

MS is a progressive autoimmune, neuroinflammatory, and neurodegenerative disease of the central nervous system (CNS), resulting from an autoimmune attack on myelinated axons [182]. This autoimmune attack induces inflammation, which leads to subsequent oligodendrocyte damage and demyelination in the CNS [183,184]. MS is a non-traumatic disabling disease affecting young adults [70]. The pathological hallmark of MS is perivascular inflammatory lesions that lead to demyelinating plaques [183]. The bulk of the inflammatory infiltrate is comprised of T-lymphocytes (dominated by major histocompatibility complex class 1 restricted CD8+ T-lymphocytes), along with much lower numbers of B-cells and plasma cells [185]. Hippocampal involvement, as evident in many patients with MS, is characterized by demyelination, neuronal damage, and synaptic abnormalities. Hippocampal demyelination is common and extensive in MS [186,187,188]. Cognitive decline, which occurs even in the absence of motor impairment, is also a hallmark of MS [189,190]. Patients with MS often show long-term memory impairments [191], attention deficiencies [192], and reduced information processing speeds [193]. Among these non-motor aspects of MS, depression, memory impairment, and psychosis are associated with hippocampal dysfunction [194] and disconnection from multiple brain networks [195].
Functional synaptic plasticity seems to be altered during the disease progression of MS [196] (Figure 1). Synaptic plasticity is disrupted in patients with MS, suggesting a synaptic basis for the cognitive abnormalities linked with this disorder [197,198]. Demyelinated hippocampi from patients with MS demonstrated a negative impact on the molecules involved in axonal transport, synaptic integrity and plasticity, Glu homeostasis, and learning and memory [188]. Demyelinated areas in the hippocampus also exhibited a significant decline in the number of synapses [188,199]. Differential outcomes on hippocampal synaptic plasticity in MS have been reported in a number of investigations. For instance, impaired hippocampal LTP and cognitive behavior are evident in both early [200,201,202] and late phase [201,202,203,204] of experimental autoimmune encephalomyelitis (EAE) mice, an animal model for MS. This impairment of LTP is associated with an altered assembly of NMDARs and a selective increase of interleukin-1β (IL-1β) in the hippocampus [200]. In contrast, increased LTP and blocked LTD are reported in the hippocampal CA1 region in EAE mice in the late phase [205]. These disparities may be attributed to the differences in EAE-inducted conditions, utilized animal, electrophysiological methods, and clinical severity at the time of evaluation, all of which interfere with the interpretation and comparability of the research [203,206].
During the past decades, many studies have focused on replicating and understanding the pathophysiological mechanisms underlying the altered synaptic plasticity in MS. For example, Di Filippo et al. [206] reviewed several mechanisms that influence synaptic plasticity during the course of MS using the EAE animal model. Inflammatory cells, including invading T lymphocytes [207], activated microglia [200,207], and some cytokines, such as IL-1β [200], play important roles in fluctuation in LTP and alterations in synaptic plasticity in EAE [207]. Particularly, LTP impairment is related to strong activation of the hippocampal microglia and elevated levels of IL-1β during the first acute relapse of relapsing-remitting EAE mice [200] and monophasic EAE rats [208]. IL-1β regulates the expression of ionotropic receptors such as NMDA [209,210] and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) [211], which are important in synaptic function and LTP induction and maintenance. Interestingly, the altered hippocampal synaptic plasticity (either enhanced or impaired) in the remitting phase of EAE is associated with persistent glial activation and elevated levels of IL-1β and nicotinamide adenine dinucleotide phosphate oxidase, a reactive oxygen species (ROS) producing enzyme [202,205,212]. Collectively, these findings imply that hippocampal LTP in EAE, and thus, in MS, is a dynamic process that is underlined by inflammatory signaling and neurotransmission (Figure 1). The onset of disturbance in LTP is debatable, although most studies demonstrate onset during late-phase rather than the early phase. A predominant hypothesis is that in comparison with the less severe early phase of the disease progression, the late phase exacerbates axonal loss, gliosis, and tissue degeneration [203,213].

2.5. Other Neurodegenerative Diseases

Apart from the above-mentioned major neurodegenerative diseases, other neurodegenerative conditions are also reported to involve the hippocampus. Both VD and FTD involve hippocampal dysfunction at variable levels [214,215]. VD is caused by cerebrovascular changes leading to cognitive impairment [216,217]. FTD is a clinically, pathologically, and genetically heterogeneous neurodegenerative disease [218]. The involvement of the hippocampus in FTD has only recently been explored [219]. Another neurodegenerative disease that affects multiple brain regions including frontotemporal, subcortical, and cerebellar regions, is ALS. Originally being a motor neuron disease [71], the hippocampal dysfunction in ALS has only recently been recognized along with memory deficits [72,220].
Synaptic dysfunction in VD has been reported in terms of decreased synaptic proteins [221,222,223], synaptic vesicles [224], and LTP [224,225] (Figure 1). In rat models of VD with bilateral common carotid artery occlusion (BCCAO), impaired basal synaptic transmission [225] and damaged synaptic ultrastructure [222] were observed. Most plasticity changes were alleviated with antioxidative therapies [65,225], supporting the strong correlation between hippocampal synaptic dysfunction and oxidative stress in VD [226]. FTD and ALS are discussed for their common pathogenic mechanisms that have different outcomes [227]. Therefore, several genetic models are subjected to experiment the common pathological outcomes in ALS and FTD. Disruption of hippocampal synaptic plasticity is reported in C9orf72-deficient mice in terms of reduced LTP and LTD [228]. Furthermore, amplitude of the Glu currents were reduced without changing the paired pulse in the hippocampal slices from UBQLN2P497H TG mice [227]. Although ALS is primarily a motor neuron disease, it is now being recognized as a multi-system condition with disease-specific patterns of hippocampal pathology [229]. The evidence for memory deficits related to hippocampal dysfunction is widely found in ALS [229,230,231,232]. Decreased functional connectivity among bilateral hippocampus, bilateral para hippocampal gyri, and cerebellum was revealed in patients with ALS [231,233]. In SOD1G93A TG mice, a genetic model for ALS, LTP impairment and decreased NMDAR-mediated synaptic currents were associated with the symptomatic stages. However, in the pre-symptomatic stage, the pre-synaptic function was enhanced, while increased adenosine A2A receptor levels were found at both symptomatic and pre-symptomatic stages [234]. Similarly, increased LTP in pre-symptomatic SOD1G93A TG mice was observed along with higher expression of GluR1 [235].
Chronic cerebral hypoperfusion due to various reasons results in glucose and oxygen depletion in the CNS, leading to the pathophysiological process of VD (Figure 1), which includes oxidative stress, inflammatory response, cell apoptosis, autophagy, and synaptic damage [236,237,238,239]. Although the precise mechanism for hippocampal synaptic dysfunction is unclear, the previously mentioned pathophysiological processes are suggested as the primary causes for the synaptic ultrastructural changes, synaptic loss, and reduction in PSD thickness [224]. Furthermore, the influence of VD-induced cholinergic deficits on LTP impairment has been described [240,241,242]. Hippocampal synaptic function in SOD1G93A TG mice, a model of ALS, is largely associated with neurotransmitter imbalances (Figure 1). For example, increased LTP in pre-symptomatic SOD1G93A TG mice was observed together with higher expression of GluR1 [235] and adenosine A2A receptor levels [234]. Adenosine A2A receptor activation enhances neuronal excitability, resulting in excitotoxic and neuroinflammatory consequences, both of which are hallmarks of ALS [243]. However, the observation of higher adenosine A2A receptor levels even at the symptomatic stage of SOD1G93A TG mice [234] with impaired LTP is still unexplained. In contrast, the NMDAR dysfunction and LTP impairment are possibly a result of an interplay between neurotransmitter systems; NMDARs are under the control of adenosine A2A receptors, during synaptic activity regulation [244]. ALS and FTD are neuropathologically related by the RNA-binding protein TDP-43 (TAR DNA-binding protein) [245]. TDP-43 is involved in the splicing, transport, and stability of several mRNAs which encode proteins playing important roles in synaptic function [246,247,248]. Thus, the genetic regulation of hippocampal synaptic plasticity in ALS and FTD cannot be excluded (Figure 1). However, hippocampal synaptic alterations in FTD are still poorly documented and need further investigation.

3. Structural Plasticity in the Hippocampus: General Overview

3.1. Dendritic Complexity

The hippocampus harbors a heterogeneous population of cells distributed in subfields DG, CA1, CA2, and CA3 [249,250]. Pyramidal neurons in the CA region consist of apical dendrites, which rise from the upper pole of the cell body, and basal dendrites, which emerge from the base of the soma to form the basal dendritic arbors [251]. The granule cells of the DG region consist of a cone-shaped apical dendritic tree emerging from an elliptical cell body [252]. Dendritic morphogenesis is a well-organized, but a complicated process that includes the formation of dendritic branches and spines, which facilitates the connection of neurons [253,254].
Dendritic arbors are highly dynamic structures that expand and retract in response to stimuli and are stabilized and maintained by post-synaptic signaling [255,256]. Structural dynamics of dendrites in normal, as well as diseased brain, include active growth and turnover of dendritic branches [256]. Such alterations in the dendritic structure include changes to the dendritic branching pattern, fragmentation of dendrites, and retraction of dendritic branches, all of which are associated with the reorganization of the neuronal cytoskeleton [257]. Microtubule dynamics play a key role in shaping the dendritic arbors. Dendrite branching and microtubule dynamics are regulated through a large number of cellular factors, including microtubule regulatory proteins [258], neurotransmitters [259], glucocorticoids [260], and various growth factors [261]. Thus, changes in any of these factors can result in alterations of the dendritic arbors, as evident in many neurological conditions [262,263,264,265].

3.2. Dendritic Spine

Hippocampal pyramidal and granule cells bear dendritic spines with minor differences in neck diameter, spine length, spine volume, head volume, and PSD area [266]. In general, dendritic spines are tiny, actin-rich, and specialized structures, which arise from the neuronal dendrites [267]. These membrane protrusions on the shaft of dendrites form the post-synaptic component of the excitatory synapse, and their structure and density are important factors in the functional plasticity [268]. Dendritic spines are morphologically categorized as mushroom, thin, stubby, filopodium, and branched [269,270]. Among them, mushroom spines are most stable and make functionally stronger synapses, which are responsible for the memory storage [271]. However, thin spines with a smaller head and narrow neck are relatively less stable [31]. Stubby spines without an obvious constriction between the head and shaft appear more stable and persistent than thin spines [272]. Filopodium spines are hair-like [273] and often short-lived [274], but are important for the rapid selecting potential of synaptic partners before a mature synapse is established [274]. Branched spines consist of multiple head regions, where each head synapses with a different pre-synaptic axon [275].
Overall, dendritic spines consist of a major component of the synapses between hippocampal neurons [276] and undergo structural plasticity in terms of formation, shedding expansion, and atrophy [277]. Rapid modifications in dendritic spines are also involved in rearrangements of the actin cytoskeleton [266]. Thus, dendritic spines serve as primary locations for synaptic function, and their disturbances can alter spine morphology and functions, as observed in normal aging as well as many neurodegenerative diseases [278].

4. Hippocampal Structural Plasticity in Neurodegenerative Diseases

Table 1 summarizes recent evidence regarding alterations of structural plasticity in the hippocampus in neurodegenerative diseases, including AD, PD, HD, MS, VD, FTD, and ALS.

4.1. Structural Plasticity in the Hippocampus with AD

4.1.1. Dendritic Complexity

Dendritic abnormalities are common in AD and appear early in the disease, which includes dystrophic neurites and decreased dendritic complexity [279]. A wide range of studies exploring the molecular and genetic mechanisms of AD established that molecular lesions in the asymptomatic phase are the early events that lead to neuronal damage and cognitive decline in the symptomatic phase [304,305]. Therein, Aβ aggregation and HPtau are accompanied by morphological changes, such as changes in neuronal shape, volume, and complexity. To this end, decreased dendritic length and dendritic arborization were observed in the hippocampi of patients with AD [279]. In experimental models of AD, dendritic complexity was demonstrated to be reduced in the hippocampi of amyloid precursor protein/presenilin 1 (APP/PS1)-TG mice [114,277,280], apolipoprotein E4 (APOE4)-TG mice [281], and primary cultured hippocampal neurons of miR-34c-transfected mice [282]. In contrast, an increase in dendritic complexity was reported in the N-tau-TG mice [283]. However, in TgCRND8 mice, no changes were observed in the dendritic arborization [112]. The variability in the observations may be due to differences in the animal models and the parameters used for selecting the neurons to be included in the studies (e.g., absence or presence of nearby Aβ plaques/NFTs).
The involvement of Aβ aggregations and HPtau in altered dendritic arborization in AD is well documented [277,306]. Aβ peptides within or near neurites are directly involved in synaptic disruption through mechanical factors, including mitochondrial dysfunction, oxidative stress, and calcium dysregulation [307,308]. A significantly high percentage of dendrites ending within and adjacent to NFTs are often reported to appear atrophic, which indicates substantial disruption of the cytoskeleton [277]. Another hypothesis suggests that fibrillary amyloid is detrimental, as they cause local structural disruptions and eventual neurite breakage [277]. Tau can bind to tubulin via its microtubule-binding domains [309] and regulate the microtubule dynamics. Deficits in tau protein led to reduced neuronal outgrowth disrupted axonal extension, and reduced microtubule density [310]. In AD, HPtau detaches from microtubules and aggregates into NFTs [306], which impairs the normal microtubule structure in the dendrites. However, rather than an individual effect, Aβ aggregates and HPtau interact to develop dendritic pathologies [306].

4.1.2. Dendritic Spine Density and Morphology

In general, reduced spine density and altered spine morphology are observed before the onset of reduction in neuronal density and synapses in the genetic models of AD. This is in corroboration with the impaired dendritic spine density reported in patients with AD [280]. Significant decrease in spine density was evident in APP/PS1-TG mice [114,280], APOE4-TG mice [281], CRND8-TG mice [112], 2576-TG mice [116], Aβ-infused rats [113,115], miR-34c-transfected mouse primary cultured hippocampal neurons [282], and hippocampal slices of Aβ-treated rats [117]. In contrast, an increase in spine density was reported in N-tau-TG mice [283]. Reduction in spine length was also noted in APP/PS1-TG mice [284]. Moreover, filopodia or thin dendritic spines were reduced in the miR-34c-transfected mouse primary cultured hippocampal neurons [282], CRND8-TG mice [112], and Aβ-infused rats [115]. On the other hand, a decrease in mushroom spines was reported in APP/PS1-TG [114] and APP-knock-in [285] mice.
There are several suggestions regarding spine loss in AD. It is hypothesized that mainly soluble, not fibrillary Aβ contributes to synaptic dysfunction and spine loss, which occur before plaque and tangle formation, and eventually lead to cognitive dysfunction [311]. Synapse loss is the most powerful pathological correlate of dementia found in the AD [50]. Aβ mediated synaptic failure is assumed to be caused by slightly high post-synaptic calcium concentrations and AMPAR elimination, which would reduce spine formation [312]. In addition, several studies have suggested that Glu circulation and GluR alterations due to Aβ pathology would result in spine pruning [112,115]. However, the underpinning molecular mechanisms remain uncertain. One of the proposed mechanisms is that Aβ might influence dendritic spines through the serum-inducible kinase-spine-associated rap guanosine triphosphatase-activating protein signaling pathway by influencing the synaptic integrity [313]. In addition, recent studies have considered tau proteins as key mediators of Aβ induced synaptic dysfunction and loss [311]. Several kinases, including cyclin-dependent kinase 5, Fyn, glycogen synthase kinase-3 (GSK3), and MAPK, can be inappropriately activated by Aβ, resulting in HPtau [314]. HPtau allows Fyn to be targeted to the PSD of dendritic spines, where it phosphorylates GluN2 and stabilizes its association with PSD95, increasing the excitotoxicity [278]. Additionally, detachment of HPtau from microtubules and its aggregations into NFTs can directly affect the actin cytoskeleton of spines [306]. Collectively, spine alterations in AD are mediated by the interplay of several factors including pathologic protein deposits, neurotransmitters, and kinases. Future studies are warranted to help elucidate the exact mechanism of this interaction. The mechanisms underlying the current AD therapy [315,316,317], which target these abnormal proteins, can be connected to the ability to rescue impairment of structural plasticity in the hippocampus with AD. Future understanding of molecular mechanisms responsible for this interplay may expose novel therapeutic targets as well as increase the effectiveness of existing anti-HPtau and Aβ therapies.

4.2. Structural Plasticity in the Hippocampus with PD

4.2.1. Dendritic Complexity

A wide range of clinical and preclinical studies of Parkinsonism have revealed dystrophic alterations in the dendrites of striatal medium spiny neurons (MSNs) [318,319,320,321,322]. However, the evidence for structural plasticity in the parkinsonian hippocampus is limited. Only a few studies have investigated hippocampal structural plasticity in animal models for PD. To the best of our knowledge, there are no such reports from clinical studies. Dendritic length and branching were reduced in primary cultured hippocampal neurons [286] and newly generated DG neurons [287,288] in the LRRK2-mutant mice. However, a previous study showed that the dendritic complexity of both CA1 and DG subregions was not altered in the MPTP-lesioned mice [289]. Therefore, further studies are needed to confirm dendritic pathology in PD animal models, since variations in observations due to different models cannot be neglected.
A previous study hypothesized that LRRK2 mutation impairs rearrangement of actin cytoskeleton in neuronal morphogenesis of the hippocampus; however, relevant mechanisms are yet to be fully elucidated [288]. In this regard, hippocampal dendritic arbors need to be investigated in PD. First, similar to the observed striatal dendritic pathology, DA is also important in the modulation of hippocampus-dependent learning and memory [323], thus interactions between DA depletion and hippocampal dysfunction in PD is highly likely [129,324]. Dopamine D1 receptors play a prominent role in synaptic plasticity and learning and memory in the hippocampus [325]. D1 receptors-mediated dendritic growth is reported in the medial prefrontal cortex [326], although it is not yet tested in the hippocampus. Second, α-Syn pathology, a hallmark of Lewy neurites in sporadic PD, impairs dendritic branching in adult-born granule cells [327] and midbrain neurons [328]. Histological evaluations revealed α-Syn as the main component that leads to axonal and dendritic pathologies in the hippocampus [329]. However, the possible effects of α-Syn pathology on dendritic arbors in the hippocampus need further clarifications using clinical and preclinical models of PD. Additionally, hippocampal serotonin and NA deficiencies were noted in several animal models for PD [148,330,331], and both serotonin and NA were involved in the regulation of the neurite extension [332]. Collectively, these findings may reveal possible consequences on hippocampal dendritic pathologies in PD.

4.2.2. Dendritic Spine Density and Morphology

In general, decreased spine density and altered morphology are described in hippocampal neurons in PD models. Spine density in the hippocampal DG region was decreased in MPTP-lesioned mice along with a remarkable decrease in the proteins that mediated neuroplasticity mechanisms and impaired cognitive function [139]. Similarly, a recent study found decreased spine density in both CA1 and DG subregions of the MPTP-lesioned mice during the late phase of PD [289]. In LRRK2-mutant mice, newly generated DG neurons were found with reduced dendritic spines [288]. Moreover, PFF-treated primary cultured hippocampal neurons exhibited reductions in dendritic spine density and head diameter of mushroom-shaped spines [290,291]. These observations suggest that the pathology of PD is not merely restricted to the nigrostriatal pathway.
There are several possible factors underlying the spine pathologies in hippocampal neurons in the PD brain, such as alterations in neurotransmission (i.e., DA, serotonergic, NA, and cholinergic systems) and Lewy bodies [333]. In mouse primary cultured hippocampal neurons, NA and DA agonists enhanced the spine density by means of elevated cAMP concentration [334], which suggests a possible mechanism underlying the spine pathology in DA-deprived hippocampi of PD. The DA levels decreased in the hippocampi of a PD animal model [324]. Similarly, deficiencies in serotonin and NA have been found in other PD animal models [148,330,331], both of which are also involved in the regulation of spine growth in hippocampal neurons [335].
Accumulation of α-Syn also occurs in the hippocampus [336]. Reductions in spine density have occurred only in wild-type neurons, but not in hippocampal neurons of α-Syn deficient mice, suggesting that the changes in spine morphology result from fibril-induced corruption of endogenously expressed α-Syn [290]. α-Syn, a protein abundant in the pre-synaptic terminals, has been postulated to polymerize purified tubulin into microtubules [337] and aid in vesicle fusion and recycling [338]. In PD, aberrant soluble oligomeric conformations of α-Syn mediate the disruption of cellular homeostasis and eventual death [339]. Additionally, α-Syn interacts with serotonin transporter activity by sequestrating it from the cellular membrane [340], which exerts deleterious effects on the structural plasticity [341]. Thus, dysregulations of dendritic spines in the hippocampus of the PD brain seem to be associated with interactions between several factors (neurotransmitters and abnormal protein aggregates), which contributes to the pathophysiology in both preclinical and clinical PD.
Observations from patients with PD suggest that neuronal changes in the hippocampus may trigger neuropsychological symptoms of PD [342]. Moreover, unlike the well-known motor functions, the neuropsychological symptoms have a relatively poor response to DA therapy, which further confirms the extranigral control of such symptoms [73]. Future studies on the alteration of neuronal architecture in the hippocampus may explain most of the unexplained pathophysiology of neuropsychological features in PD.

4.3. Structural Plasticity in the Hippocampus with HD

4.3.1. Dendritic Complexity

Impairments in the dendritic complexity have been well documented in the MSNs of the striatum in animal models of HD [343,344,345]. However, dendritic structure in hippocampal neurons widely remains unknown in HD patients and animal models. A study using primary hippocampal neurons from R6/1-TG mice reported a decrease in neurite number and complexity [292]. However, the evidence was insufficient to draw conclusions about hippocampal dendritic changes in HD. Therefore, the description of hippocampal structural plasticity in HD pathogenesis requires further elucidation in both clinical and preclinical studies.
The pathology of aberrant dendritic structure in HD has been linked to the involvement of the mutant huntingtin gene (mhtt) [346]. In the normal brain, htt is an important regulator of mitochondrial function, whereas in the mutant background, mhtt causes several mitochondrial changes, such as loss of membrane potential and increased oxidative stress [346]. mhtt affects ATP levels in synapses [347] and leads to possible pathophysiological implications since several molecular processes in the synapses are susceptible to low ATP levels [348]. Hippocampal dendrite outgrowth is a highly energy-dependent process [349], which may be impaired when there is a reduction in mitochondrial ATP production [350]. Recent developments in metabolic research at the cellular level have provided critical insights into the cellular and molecular underpinnings of the connection between neural activity and energy consumption [351]. These techniques may help in finding methods for restoring ATP in synapses with mhtt-induced low ATP levels. Additionally, mhtt is also involved in the vesicular transport of neurotrophic molecules like BDNF along the microtubules [352], which are important for dendritic tree development. Ultimately, the functional capacity of the hippocampus is directly related to the complexity of neuronal dendritic arbors [353]. In addition, stimulation of prostaglandin E2 receptors increased the dendritic growth, recovered the neurochemical levels in R6/1-TG primary hippocampal neurons, and reduced mhtt-induced cognitive deficits in R6/1-TG mice, suggesting its therapeutic potential for HD [292].

4.3.2. Dendritic Spine Density and Morphology

There are conflicting findings of alterations of dendritic spines in hippocampal neurons of the HD brain. Reduction of dendritic spine density in the CA1 subregion of the hippocampus has been reported in Hip14-deficient [293], R6/1-TG [294], and Pyk2-deficient [172] mice. However, in a different study that used R6/1-TG mice, dendritic spine density in the hippocampus remained unchanged [295]. These discrepancies in observations may be due to differences in the experimental setup and genetic background. Structural changes in hippocampal spines are frequently synonymous with impaired learning and memory in animal models of HD [172,292,293,294]. Therefore, the structural changes in hippocampal dendritic spines may have a role in the cognitive and neuropsychiatric deficits in HD.
The underlying mechanism for spine pathology in HD is still not clear. mhtt affects a variety of cellular and molecular processes that influence loss of spines and synaptic dysfunction, such as protein trafficking and aggregation, protein-protein interaction, calcium signaling, mitochondrial function, gene transcription, neurotransmitter release, and receptor activation, and neurotrophic support [347,354,355]. Among them, the mitochondrial function appears to be the predominant factor affecting spine maturation and morphogenesis [356]. For instance, in hippocampal neurons, manipulations that reduce dendritic mitochondrial content led to loss of synapses and dendritic spines [357]. mhtt impaired the mitochondrial transport in neuronal processes of the hippocampus of BACHD-TG mice [358], suggesting a possible mechanism involving spine pathologies in HD. Chronic inflammation involving glial activation is another possible pathway that can negatively impact hippocampal spine density in HD. For example, localized inflammatory response in the CA1 region and upregulation of nuclear factor kappa B (NF-κB) signaling have been observed in R6/1-TG mice along with reduced spine density [294]. Treatments to alleviate inflammatory responses also recovered these spine pathologies [359]. Thus, the use of anti-inflammatory pathways in maintaining spine structural integrity in HD is important.

4.4. Structural Plasticity in the Hippocampus with MS

4.4.1. Dendritic Complexity

Aberrant neurite orientation dispersion in the spinal cord serves as a marker of microstructural pathology in patients with MS [360]. Extensive dendrite and neuronal soma atrophy have also been observed following axonal lesions in the spinal cord of patients with MS [361]. However, hippocampal dendritic pathologies in MS are also reported. In EAE mice at 20 days post-induction, dendritic length, and intersections were reduced in the molecular layer of the hippocampal DG [296,297]. These structural changes coexisted with hippocampus-dependent memory deficits in the mice [297].
In MS, there are several possible explanations for these structural modifications in the hippocampus. T-cell mediated autoimmunity and reactive gliosis trigger inflammatory mediators, including tumor necrosis factor α (TNFα) and interferon γ (IFNγ), which are toxic to oligodendrocytes [362,363,364] and inhibit neuronal precursor cell proliferation [365]. Defects in oligodendrocyte proliferation lead to hippocampal demyelination, which in turn leads to decreased dendritic arborization of hippocampal neurons [366]. The neurons in the CA1 region are highly vulnerable to a variety of stressors, including glutamate-mediated excitotoxicity [367], which has been implicated in the MS pathogenesis [368]. Another potential factor that may influence hippocampal neuroarchitecture is high-dose corticosteroids, which are routinely used to treat acute relapses. Corticosteroids significantly affect the CA1 neurons [369,370]. In fact, patients with MS, who are given large doses of steroids, experience transitory abnormalities in the declarative memory [371,372,373]. Thus, morphological changes in neurons of the hippocampus could reflect a variety of possible neuropathological processes, including demyelination, decreased dendritic density, and/or neuronal loss. However, the presently available evidence is too limited to attain a consensus regarding the role of hippocampal structural plasticity in MS.

4.4.2. Dendritic Spine Density and Morphology

Dendritic spine alterations in the hippocampus have not been evaluated in patients with MS as well as EAE mice. Particularly, as fatality is rare in the early stage of MS, there is limited information about the acute effects of hippocampal demyelination on spine pathologies in patients with MS. However, there was no change in the spine density of demyelinated hippocampi of cuprizone-diet fed mice, except for an increase in the proportion of mushroom-shaped spines [298]. This increase was nullified with the withdrawal of the cuprizone diet and subsequent remyelination.
In the cuprizone-diet fed mice, the precise mechanism that links demyelination to dendritic spine abnormalities remains speculative. One possibility is that an inflammatory response via microglial activation may decrease the synaptic connectivity during the cuprizone-diet-induced demyelination [374]. Second, it has been shown that activated microglia secrete inflammatory cytokines and/or chemokines (i.e., IL-1β, TNFα, and IFNγ), which may also negatively impact neurogenesis and modulate dendritic spine morphology [375,376]. Third, as brain activity is a critical regulator of neuronal proliferation and synaptic connectivity [377,378], reduced brain activity due to demyelination can negatively impact hippocampal neurogenesis and spine densities. Nevertheless, the direct effect of cuprizone on proliferating cells and synaptic connectivity cannot be ignored [375]. However, the current findings from cuprizone-diet-induced demyelination in the hippocampus are not enough to infer its relevance to MS. Moreover, the demyelination in MS is more likely to occur over decades (chronic) in contrast to the acute demyelination mediated by the cuprizone diet. Therefore, a complete elucidation of the effect of demyelination on spine pathology in animal models with chronic hippocampal demyelination is warranted.
The association between microglial activation, demyelination, neurodegeneration, and disability has been established in patients with MS [379] as well as in EAE animal models [380]. Planche et al. [297] suggested a relationship between microglial activation, dendritic damages, synaptic plasticity disruption, and memory impairment, especially in the DG region. Despite being the most widely used model of MS, the specific spine pathologies in the hippocampus of EAE models remain to be elucidated.

4.5. Structural Plasticity in the Hippocampus with Other Neurodegenerative Diseases

4.5.1. Dendritic Complexity

Total dendritic length, dendrite numbers, and dendrite crossings were significantly reduced in both CA1 and DG regions of rat models of VD with BCCAO [65,299]. Hippocampal dendritic branching and outgrowth were reduced in primary cultured rat hippocampal neurons with TDP-43 overexpression, as in an in vitro model of ALS/FTD [67]. In hSOD1G93A TG mice, aberrant dendritic complexity is readily reported in motor neurons of the primary motor cortex [381], pyramidal neurons of the medial prefrontal cortex [382], and lower motor neurons of the brainstem and spinal cord [383,384]. However, no changes were observed in the hippocampal dendritic complexity in the SOD1G93A TG mice [303].
Chronic brain hypoperfusion induces Aβ aggregation, HPtau, and cell death [385,386,387]. In VD rats, aberrant dendritic complexity in the hippocampus was associated with miR-195 mediated overproduction of N-terminal β-amyloid precursor protein (N-APP) [299], suggesting that miR-195 is a key link between the hallmarks of both AD and VD. It is well known that Aβ aggregation insults the dendritic complexity of hippocampal neurons, as described in Section 4.1.1. Therefore, reduction of hippocampal dendritic complexity in VD would share, at least in part, the same pathological mechanisms with AD. In addition, as one of the risk factors for VD, arterial hypertension has recently been reviewed as an altering factor of the hippocampal dendritic complexity in both human and animal models [388]. In a proposed model for ALS/FTD, dendritic growth is suppressed with the elevation of TDP-43 protein [67], primarily through RNA-binding functions that disrupt the functionality of many RNA transcripts. Furthermore, microtubule-associated tau (MAPT) is one of the three most common gene mutations in the FTD [389]. MAPT is critical in the formation and stabilization of axonal microtubules and neurite growth, and its mutation changes dendritic structure [390,391]. However, changes in hippocampal structural plasticity in ALS and FTD are still unclear due to the limited and contradictory literature arising from different animal models.

4.5.2. Dendritic Spine Density and Morphology

In VD rats, densities of dendritic spines in both DG [65,300] and CA1 [301,302] regions of the hippocampus were decreased, largely due to the decrease in mushroom spines [65,301,305]. Moreover, expressions of spinophilin, a spine marker protein [301], and BDNF [302] were significantly lower in the CA1 region of VD rats. Similarly, with the major neurodegenerative diseases, aberrant hippocampal spine density was also associated with cognitive deficits in VD [301,302]. However, there was no difference in dendritic spine density in the DG region of the hippocampus of SOD1G93A TG mice, an animal model for ALS [303].
In general, cerebral hypoperfusion in VD animal models results in depleted ATP, mitochondrial dysfunction, and thereby increased ROS production and oxidative stress leading to a range of pathological events [392]. However, several molecules that play key roles in the major neurodegenerative diseases seem to take part in non-AD dementia as well. BDNF is one neurotrophic factor of such importance, which was found to be decreased in the VD hippocampus together with the suppression of the AMPK pathway [302]. Enhancement of BDNF levels and activation of the AMPK pathway in the hippocampus with VD simultaneously cured the spine pathology and cognitive deficits [302]. However, the role of the AMPK pathway in VD and other neurodegenerative diseases should be further studied, as there are contradictory reports on AMPK activation in neurodegenerative diseases, reporting worsening of the neuropathological and behavioral phenotypes [393,394]. Another highly plausible pathway, which could be involved in the spine pathology observed in VD, is GSK-3β and β-catenin signaling. GSK-3β activation has been noted in an animal model for VD in parallel with hippocampal spine density reduction [300]. Activation of GSK-3β has also been shown to increase the levels of pro-inflammatory cytokines, such as IL-1β and TNF-α [395], which further leads to detrimental effects on the dendritic structural plasticity [396,397]. Thus, the interplay of these proposed mechanisms needs further studies in order to clarify their role in the dendritic spine pathologies observed in VD. Moreover, alteration of spine density and morphology in other neurodegenerative diseases, such as FTD and ALS, are poorly documented and need further investigations.
Figure 2 illustrates the plausible mechanistic pathways underpinning the alterations of hippocampal structural plasticity in neurodegenerative diseases. The majority of previous studies demonstrate that pathological molecules (Aβ plaques, Tau proteins, and α-Syn), mutated genes (mhtt), and neuroinflammation underpin the structural pathologies in hippocampal neurons. Therefore, targeted therapies against pathological molecules, mutated genes, and neuroinflammation, which restore hippocampal structural pathologies, may offer a novel mechanism to alleviate the symptoms of neurodegenerative diseases.

5. Hippocampal Dysfunction and Structural Plasticity in the Aging Brain

Apart from the neurodegenerative conditions, hippocampal dysfunction and cognitive impairment are often associated with aging [398,399,400,401]. Age-related hippocampal dysfunction is a broad topic and a major risk factor for neurodegenerative diseases, such as AD [402]. Several recent review articles offer excellent information on the involvement of hippocampal dysfunction in the context of aging [402,403,404,405,406].
Age-dependent aberrations of hippocampal dendritic complexity are well documented [407,408], with older age being directly correlated with impairments in dendritic complexity. Sex-related factors include female mice being less susceptible to aging-related dendritic complexity, at least in the CA1 region [409]. The influence of sex hormones and aging on hippocampal structural plasticity is adeptly discussed by Galea et al. [56].
Moreover, many studies described decreased dendritic spine density in aged mice hippocampi [407,408]. In contrast, another research group described unchanged spine density in the hippocampus over aging [409]. However, the influence of other factors like sex hormones cannot be excluded in these incongruent observations [56]. Detailed regional susceptibilities in aging-related dendritic spine pathology in the hippocampus have already been discussed previously [57]. Collectively, complex heterogeneous neurodegenerative mechanisms are presumably involved in alterations of hippocampal structural plasticity in aging brains.

6. Concluding Remarks

The hippocampus is one of the brain areas that shows neuronal remodeling in many neurodegenerative diseases, including AD, PD, HD, and MS. Neuronal remodeling proceeds through structural changes in the dendrites and spines of hippocampal neurons. The current review presented an overview of structural modifications of the hippocampus in neurodegenerative diseases, along with the underlying mechanisms. This review confirmed that hippocampal neuroarchitecture is affected at different levels (functional and structural) and to varying degrees in these neurodegenerative diseases.
In AD, alterations in dendritic complexity and spine density of hippocampal neurons start appearing in the early phase of disease progression. Moreover, the main molecular cues underlying these morphological changes in hippocampal neurons are Aβ plaques and HPtau, which alter the microtubule arrangement in the cells. The majority of the studies regarding PD elaborate on structural impairment in the context of striatal MSNs. Nevertheless, a few recent studies have shifted the focus to the hippocampus. In HD, dendritic spine loss in CA1 is the main neuroplasticity change in the hippocampus. Up-to-date evidence suggests that mhtt is responsible for these observed structural changes in HD, which is caused by impaired synaptic energy metabolism caused by decreased mitochondrial function. As the dendritic growth is highly sensitive to the supply of ATP, the impaired energy metabolism in HD may influence neuronal dendritic arbor formation. In MS, alterations in structural plasticity led to severe demyelination and synaptic abnormalities in the hippocampus. Activated inflammatory cells and impaired brain activity could be the main factors responsible for the changes in dendritic arborization and spine density in the hippocampi of patients with MS and animal models. In VD, inflammation and the activation of kinases upon chronic hypoperfusion are associated with structural changes in both dendritic arbors and spines. However, available literature on FTD and ALS is insufficient to make judgments on structural plasticity changes in these diseases. Collectively, different aspects of hippocampal structural plasticity were greatly affected in neurodegenerative diseases that are discussed in the current review. The negative changes in structural plasticity in the hippocampus are positively correlated with disease severity and further impact the functional symptoms in neurodegenerative diseases. However, there are still many gaps in understanding how the functional and structural changes of hippocampal neurons occur in these neurodegenerative diseases.
Consequently, this review provided an update on the status of the understanding of the role of hippocampal structural plasticity in the etiopathogenesis of neurodegenerative diseases. The current focus and existing gaps in the knowledge of hippocampal neuroplasticity in neurodegenerative diseases are also identified in this review. Conclusively, the information presented in this review may serve as a starting point for future mechanistic and therapeutic research in the field.

Author Contributions

Conceptualization, C.M.; methodology, M.J.A., P.D.E.W.-M., S.K., J.-S.K. and C.M.; investigation, M.J.A., P.D.E.W.-M., S.K., J.-S.K. and C.M.; resources, C.M.; data curation, M.J.A. and P.D.E.W.-M.; visualization, M.J.A. and P.D.E.W.-M.; writing—original draft preparation, P.D.E.W.-M., M.J.A. and C.M.; writing–review and editing, M.J.A., P.D.E.W.-M. and C.M.; supervision, C.M.; project administration, C.M.; funding acquisition, C.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation (NRF) of Korea grant funded by the Korean Government (NRF-2020R1A4A1019395; NRF-2022R1A2C1004022).

Data Availability Statement

This paper utilized original data not used in other publications. The datasets generated and/or analyzed in the present study are available from the corresponding author upon reasonable request.

Acknowledgments

Due to the space limit, we apologize that this review may have missed some of the relevant literature.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schaefers, A.T.; Teuchert-Noodt, G. Developmental neuroplasticity and the origin of neurodegenerative diseases. World J. Biol. Psychiatry 2016, 17, 587–599. [Google Scholar] [CrossRef] [PubMed]
  2. Keller, T.A.; Just, M.A. Structural and functional neuroplasticity in human learning of spatial routes. Neuroimage 2016, 125, 256–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sala, C.; Segal, M. Dendritic spines: The locus of structural and functional plasticity. Physiol. Rev. 2014, 94, 141–188. [Google Scholar] [CrossRef]
  4. Das, S.; Sadanandappa, M.K.; Dervan, A.; Larkin, A.; Lee, J.A.; Sudhakaran, I.P.; Priya, R.; Heidari, R.; Holohan, E.E.; Pimentel, A.; et al. Plasticity of local GABAergic interneurons drives olfactory habituation. Proc. Natl. Acad. Sci. USA 2011, 108, E646–E654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Yuan, Q.; Xiang, Y.; Yan, Z.; Han, C.; Jan, L.Y.; Jan, Y.N. Light-induced structural and functional plasticity in Drosophila larval visual system. Science 2011, 333, 1458–1462. [Google Scholar] [CrossRef] [Green Version]
  6. Hotulainen, P.; Hoogenraad, C.C. Actin in dendritic spines: Connecting dynamics to function. J. Cell Biol. 2010, 189, 619–629. [Google Scholar] [CrossRef] [Green Version]
  7. Luo, L. Actin cytoskeleton regulation in neuronal morphogenesis and structural plasticity. Annu. Rev. Cell Dev. Biol. 2002, 18, 601–635. [Google Scholar] [CrossRef] [Green Version]
  8. Peche, V.; Shekar, S.; Leichter, M.; Korte, H.; Schröder, R.; Schleicher, M.; Holak, T.; Clemen, C.; Ramanath-y, B.; Pfitzer, G. CAP2, cyclase-associated protein 2, is a dual compartment protein. Cell. Mol. Life Sci. 2007, 64, 2702–2715. [Google Scholar] [CrossRef]
  9. Lai, K.O.; Ip, N.Y. Structural plasticity of dendritic spines: The underlying mechanisms and its dysregulation in brain disorders. Biochim. Biophys. Acta 2013, 1832, 2257–2263. [Google Scholar] [CrossRef] [Green Version]
  10. Dent, E.W.; Gertler, F.B. Cytoskeletal dynamics and transport in growth cone motility and axon guidance. Neuron 2003, 40, 209–227. [Google Scholar] [CrossRef] [Green Version]
  11. Van Aelst, L.; Cline, H.T. Rho GTPases and activity-dependent dendrite development. Curr. Opin. Neurobiol. 2004, 14, 297–304. [Google Scholar] [CrossRef] [PubMed]
  12. Verpelli, C.; Piccoli, G.; Zibetti, C.; Zanchi, A.; Gardoni, F.; Huang, K.; Brambilla, D.; Di Luca, M.; Battaglioli, E.; Sala, C. Synaptic activity controls dendritic spine morphology by modulating eEF2-dependent BDNF synthesis. J. Neurosci. 2010, 30, 5830–5842. [Google Scholar] [CrossRef] [PubMed]
  13. Rex, C.S.; Lin, C.Y.; Kramar, E.A.; Chen, L.Y.; Gall, C.M.; Lynch, G. Brain-derived neurotrophic factor promotes long-term potentiation-related cytoskeletal changes in adult hippocampus. J. Neurosci. 2007, 27, 3017–3029. [Google Scholar] [CrossRef] [PubMed]
  14. Vanderklish, P.W.; Edelman, G.M. Dendritic spines elongate after stimulation of group 1 metabotropic glutamate receptors in cultured hippocampal neurons. Proc. Natl. Acad. Sci. USA 2002, 99, 1639–1644. [Google Scholar] [CrossRef] [Green Version]
  15. Kutsarova, E.; Schohl, A.; Munz, M.; Wang, A.; Zhang, Y.Y.; Bilash, O.M.; Ruthazer, E.S. BDNF signaling in Hebbian and Stentian structural plasticity in the developing visual system. BioRxiv 2021. [Google Scholar] [CrossRef]
  16. Mattson, M.P. Glutamate and neurotrophic factors in neuronal plasticity and disease. Ann. N. Y. Acad. Sci. 2008, 1144, 97–112. [Google Scholar] [CrossRef] [Green Version]
  17. Xu, J.; Sriramula, S.; Lazartigues, E. Excessive Glutamate Stimulation Impairs ACE2 Activity Through ADAM17-Mediated Shedding in Cultured Cortical Neurons. Cell Mol. Neurobiol. 2018, 38, 1235–1243. [Google Scholar] [CrossRef]
  18. Gordon-Weeks, P.R.; Fournier, A.E. Neuronal cytoskeleton in synaptic plasticity and regeneration. J. Neurochem. 2014, 129, 206–212. [Google Scholar] [CrossRef]
  19. Hu, X.; Ballo, L.; Pietila, L.; Viesselmann, C.; Ballweg, J.; Lumbard, D.; Stevenson, M.; Merriam, E.; Dent, E.W. BDNF-induced increase of PSD-95 in dendritic spines requires dynamic microtubule invasions. J. Neurosci. 2011, 31, 15597–15603. [Google Scholar] [CrossRef] [Green Version]
  20. Ho, V.M.; Lee, J.A.; Martin, K.C. The cell biology of synaptic plasticity. Science 2011, 334, 623–628. [Google Scholar] [CrossRef] [Green Version]
  21. Mainardi, M.; Fusco, S.; Grassi, C. Modulation of hippocampal neural plasticity by glucose-related signaling. Neural. Plast. 2015, 2015, 657928. [Google Scholar] [CrossRef] [PubMed]
  22. Yamazaki, Y.; Fujiwara, H.; Kaneko, K.; Hozumi, Y.; Xu, M.; Ikenaka, K.; Fujii, S.; Tanaka, K.F. Short- and long-term functional plasticity of white matter induced by oligodendrocyte depolarization in the hippocampus. Glia 2014, 62, 1299–1312. [Google Scholar] [CrossRef] [PubMed]
  23. Magee, J.C.; Grienberger, C. Synaptic Plasticity Forms and Functions. Annu. Rev. Neurosci. 2020, 43, 95–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Citri, A.; Malenka, R.C. Synaptic plasticity: Multiple forms, functions, and mechanisms. Neuropsychopharmacology 2008, 33, 18–41. [Google Scholar] [CrossRef] [Green Version]
  25. Ren, K.; Dubner, R. Pain facilitation and activity-dependent plasticity in pain modulatory circuitry: Role of BDNF-TrkB signaling and NMDA receptors. Mol. Neurobiol. 2007, 35, 224–235. [Google Scholar] [CrossRef] [PubMed]
  26. Ciccarelli, A.; Giustetto, M. Role of ERK signaling in activity-dependent modifications of histone proteins. Neuropharmacology 2014, 80, 34–44. [Google Scholar] [CrossRef] [PubMed]
  27. Alicea, D.; Perez, M.; Maldonado, C.; Dominicci-Cotto, C.; Marie, B. Cortactin Is a Regulator of Activity-Dependent Synaptic Plasticity Controlled by Wingless. J. Neurosci. 2017, 37, 2203–2215. [Google Scholar] [CrossRef] [Green Version]
  28. Reymann, K.G.; Frey, J.U. The late maintenance of hippocampal LTP: Requirements, phases, ‘synaptic tagging’, ‘late-associativity’ and implications. Neuropharmacology 2007, 52, 24–40. [Google Scholar] [CrossRef]
  29. Bliss, T.V.; Collingridge, G.L. A synaptic model of memory: Long-term potentiation in the hippocampus. Nature 1993, 361, 31–39. [Google Scholar] [CrossRef]
  30. Sajikumar, S.; Frey, J.U. Late-associativity, synaptic tagging, and the role of dopamine during LTP and LTD. Neurobiol. Learn. Mem. 2004, 82, 12–25. [Google Scholar] [CrossRef]
  31. Chidambaram, S.B.; Rathipriya, A.G.; Bolla, S.R.; Bhat, A.; Ray, B.; Mahalakshmi, A.M.; Manivasagam, T.; Thenmozhi, A.J.; Essa, M.M.; Guillemin, G.J.; et al. Dendritic spines: Revisiting the physiological role. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 92, 161–193. [Google Scholar] [CrossRef] [PubMed]
  32. Hillman, C.H.; Erickson, K.I.; Kramer, A.F. Be smart, exercise your heart: Exercise effects on brain and cognition. Nat. Rev. Neurosci. 2008, 9, 58–65. [Google Scholar] [CrossRef] [PubMed]
  33. Mora, F. Successful brain aging: Plasticity, environmental enrichment, and lifestyle. Dialogues Clin. Neurosci. 2013, 15, 45–52. [Google Scholar]
  34. Leuner, B.; Falduto, J.; Shors, T.J. Associative memory formation increases the observation of dendritic spines in the hippocampus. J. Neurosci. 2003, 23, 659–665. [Google Scholar] [CrossRef]
  35. Beauquis, J.; Roig, P.; De Nicola, A.F.; Saravia, F. Short-term environmental enrichment enhances adult neurogenesis, vascular network and dendritic complexity in the hippocampus of type 1 diabetic mice. PLoS ONE 2010, 5, e13993. [Google Scholar] [CrossRef]
  36. Kang, S.; Son, Y.; Lee, S.; Kim, J.; Kim, J.C.; Kim, J.S.; Jung, U.; Kim, S.H.; Yang, M.; Moon, C. Changes in epigenetic markers, DNMT1 and HDAC1/2, in the adult mouse hippocampus after cranial irradiation. Neurosci. Lett. 2017, 657, 113–119. [Google Scholar] [CrossRef]
  37. Son, Y.; Kang, S.; Kim, J.; Lee, S.; Kim, J.C.; Kim, S.H.; Kim, J.S.; Jo, S.K.; Jung, U.; Youn, B.; et al. Possible involvement of hippocampal immediate-early genes in contextual fear memory deficit induced by cranial irradiation. Neurobiol. Learn Mem. 2016, 133, 19–29. [Google Scholar] [CrossRef]
  38. Sebastian, V.; Estil, J.B.; Chen, D.; Schrott, L.M.; Serrano, P.A. Acute physiological stress promotes clustering of synaptic markers and alters spine morphology in the hippocampus. PLoS ONE 2013, 8, e79077. [Google Scholar] [CrossRef] [PubMed]
  39. Cymerman, I.A.; Gozdz, A.; Urbanska, M.; Milek, J.; Dziembowska, M.; Jaworski, J. Structural Plasticity of Dendritic Spines Requires GSK3alpha and GSK3beta. PLoS ONE 2015, 10, e0134018. [Google Scholar] [CrossRef]
  40. Pyapali, G.K.; Turner, D.A. Increased dendritic extent in hippocampal CA1 neurons from aged F344 rats. Neurobiol. Aging 1996, 17, 601–611. [Google Scholar] [CrossRef]
  41. Graham, B.P.; van Ooyen, A. Mathematical modelling and numerical simulation of the morphological development of neurons. BMC Neurosci. 2006, 7 (Suppl. 1), S9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Quackenbush, L.J.; Ngo, H.; Pentney, R.J. Evidence for nonrandom regression of dendrites of Purkinje neurons during aging. Neurobiol. Aging 1990, 11, 111–115. [Google Scholar] [CrossRef]
  43. Smith-Dijak, A.I.; Sepers, M.D.; Raymond, L.A. Alterations in synaptic function and plasticity in Huntington disease. J. Neurochem. 2019, 150, 346–365. [Google Scholar] [CrossRef] [PubMed]
  44. Nguyen, M.; Wong, Y.C.; Ysselstein, D.; Severino, A.; Krainc, D. Synaptic, Mitochondrial, and Lysosomal Dysfunction in Parkinson’s Disease. Trends Neurosci. 2019, 42, 140–149. [Google Scholar] [CrossRef] [PubMed]
  45. Herms, J.; Dorostkar, M.M. Dendritic Spine Pathology in Neurodegenerative Diseases. Annu. Rev. Pathol. 2016, 11, 221–250. [Google Scholar] [CrossRef] [PubMed]
  46. Mikkonen, M.; Soininen, H.; Alafuzof, I.; Miettinen, R. Hippocampal plasticity in Alzheimer’s disease. Rev. Neurosci. 2001, 12, 311–325. [Google Scholar] [CrossRef] [PubMed]
  47. Verkhratsky, A.; Rodriguez, J.J.; Parpura, V. Astroglia in neurological diseases. Future Neurol. 2013, 8, 149–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Luebke, J.I.; Weaver, C.M.; Rocher, A.B.; Rodriguez, A.; Crimins, J.L.; Dickstein, D.L.; Wearne, S.L.; Hof, P.R. Dendritic vulnerability in neurodegenerative disease: Insights from analyses of cortical pyramidal neurons in transgenic mouse models. Brain Struct Funct. 2010, 214, 181–199. [Google Scholar] [CrossRef] [Green Version]
  49. Kweon, J.H.; Kim, S.; Lee, S.B. The cellular basis of dendrite pathology in neurodegenerative diseases. BMB Rep. 2017, 50, 5–11. [Google Scholar] [CrossRef] [Green Version]
  50. Penzes, P.; Cahill, M.E.; Jones, K.A.; VanLeeuwen, J.E.; Woolfrey, K.M. Dendritic spine pathology in neuropsychiatric disorders. Nat. Neurosci. 2011, 14, 285–293. [Google Scholar] [CrossRef] [Green Version]
  51. Gorrie, G.H.; Fecto, F.; Radzicki, D.; Weiss, C.; Shi, Y.; Dong, H.; Zhai, H.; Fu, R.; Liu, E.; Li, S.; et al. Dendritic spinopathy in transgenic mice expressing ALS/dementia-linked mutant UBQLN2. Proc. Natl. Acad. Sci. USA 2014, 111, 14524–14529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kim, H.; Yi, J.H.; Choi, K.; Hong, S.; Shin, K.S.; Kang, S.J. Regional differences in acute corticosterone-induced dendritic remodeling in the rat brain and their behavioral consequences. BMC Neurosci. 2014, 15, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ang, M.J.; Lee, S.; Wada, M.; Weerasinghe-Mudiyanselage, P.D.E.; Kim, S.H.; Shin, T.; Jeon, T.I.; Im, S.S.; Moon, C. SREBP-1c Deficiency Affects Hippocampal Micromorphometry and Hippocampus-Dependent Memory Ability in Mice. Int. J. Mol. Sci. 2021, 22, 6103. [Google Scholar] [CrossRef]
  54. Torres, M.D.; Garcia, O.; Tang, C.; Busciglio, J. Dendritic spine pathology and thrombospondin-1 deficits in Down syndrome. Free Radic Biol. Med. 2018, 114, 10–14. [Google Scholar] [CrossRef] [PubMed]
  55. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
  56. Galea, L.A.; Leuner, B.; Slattery, D.A. Hippocampal plasticity during the peripartum period: Influence of sex steroids, stress and ageing. J. Neuroendocrinol. 2014, 26, 641–648. [Google Scholar] [CrossRef] [PubMed]
  57. Dickstein, D.L.; Weaver, C.M.; Luebke, J.I.; Hof, P.R. Dendritic spine changes associated with normal aging. Neuroscience 2013, 251, 21–32. [Google Scholar] [CrossRef] [Green Version]
  58. Boros, B.D.; Greathouse, K.M.; Gearing, M.; Herskowitz, J.H. Dendritic spine remodeling accompanies Alzheimer’s disease pathology and genetic susceptibility in cognitively normal aging. Neurobiol. Aging. 2019, 73, 92–103. [Google Scholar] [CrossRef] [PubMed]
  59. Moreno-Jimenez, E.P.; Flor-Garcia, M.; Terreros-Roncal, J.; Rabano, A.; Cafini, F.; Pallas-Bazarra, N.; Avila, J.; Llorens-Martin, M. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 2019, 25, 554–560. [Google Scholar] [CrossRef]
  60. Tada, T.; Sheng, M. Molecular mechanisms of dendritic spine morphogenesis. Curr. Opin. Neurobiol. 2006, 16, 95–101. [Google Scholar] [CrossRef]
  61. Leuner, B.; Gould, E. Structural plasticity and hippocampal function. Annu. Rev. Psychol. 2010, 61, 111–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Mateus-Pinheiro, A.; Pinto, L.; Bessa, J.M.; Morais, M.; Alves, N.D.; Monteiro, S.; Patricio, P.; Almeida, O.F.; Sousa, N. Sustained remission from depressive-like behavior depends on hippocampal neurogenesis. Transl. Psychiatry 2013, 3, e210. [Google Scholar] [CrossRef]
  63. Dioli, C.; Patricio, P.; Trindade, R.; Pinto, L.G.; Silva, J.M.; Morais, M.; Ferreiro, E.; Borges, S.; Mateus-Pinheiro, A.; Rodrigues, A.J.; et al. Tau-dependent suppression of adult neurogenesis in the stressed hippocampus. Mol. Psychiatry 2017, 22, 1110–1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sun, B.; Halabisky, B.; Zhou, Y.; Palop, J.J.; Yu, G.; Mucke, L.; Gan, L. Imbalance between GABAergic and Glutamatergic Transmission Impairs Adult Neurogenesis in an Animal Model of Alzheimer’s Disease. Cell Stem Cell 2009, 5, 624–633. [Google Scholar] [CrossRef] [Green Version]
  65. Zhu, T.; Zhu, M.; Qiu, Y.; Wu, Z.; Huang, N.; Wan, G.; Xu, J.; Song, P.; Wang, S.; Yin, Y.; et al. Puerarin Alleviates Vascular Cognitive Impairment in Vascular Dementia Rats. Front Behav. Neurosci. 2021, 15, 717008. [Google Scholar] [CrossRef] [PubMed]
  66. Moodley, K.K.; Chan, D. The hippocampus in neurodegenerative disease. Front. Neurol. Neurosci. 2014, 34, 95–108. [Google Scholar] [CrossRef]
  67. Herzog, J.J.; Deshpande, M.; Shapiro, L.; Rodal, A.A.; Paradis, S. TDP-43 misexpression causes defects in dendritic growth. Sci. Rep. 2017, 7, 15656. [Google Scholar] [CrossRef] [PubMed]
  68. Duff, K.; Paulsen, J.; Mills, J.; Beglinger, L.J.; Moser, D.J.; Smith, M.M.; Langbehn, D.; Stout, J.; Queller, S.; Harrington, D.L.; et al. Mild cognitive impairment in prediagnosed Huntington disease. Neurology 2010, 75, 500–507. [Google Scholar] [CrossRef] [Green Version]
  69. Checkoway, H.; Lundin, J.I.; Kelada, S.N. Neurodegenerative diseases. IARC Sci. Publ. 2011, 163, 407–419. [Google Scholar]
  70. Kobelt, G.; Thompson, A.; Berg, J.; Gannedahl, M.; Eriksson, J.; Group, M.S.; Platform, E.M.S. New insights into the burden and costs of multiple sclerosis in Europe. Mult. Scler. J. 2017, 23, 1123–1136. [Google Scholar] [CrossRef]
  71. Hardiman, O.; Al-Chalabi, A.; Chio, A.; Corr, E.M.; Logroscino, G.; Robberecht, W.; Shaw, P.J.; Simmons, Z.; van den Berg, L.H. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 2017, 3, 17085. [Google Scholar] [CrossRef] [PubMed]
  72. Beeldman, E.; Raaphorst, J.; Klein Twennaar, M.; de Visser, M.; Schmand, B.A.; de Haan, R.J. The cognitive profile of ALS: A systematic review and meta-analysis update. J. Neurol. Neurosurg Psychiatry 2016, 87, 611–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chaudhuri, K.R.; Healy, D.G.; Schapira, A.H.; National Institute for Clinical, E. Non-motor symptoms of Parkinson’s disease: Diagnosis and management. Lancet Neurol. 2006, 5, 235–245. [Google Scholar] [CrossRef]
  74. Estrada-Bellmann, I.; Camara-Lemarroy, C.R.; Calderon-Hernandez, H.J.; Rocha-Anaya, J.J.; Villareal-Velazquez, H.J. Non-motor symptoms and quality of life in patients with Parkinson’s disease in Northeastern Mexico. Acta Neurol. Belg. 2016, 116, 157–161. [Google Scholar] [CrossRef]
  75. Liu, W.M.; Lin, R.J.; Yu, R.L.; Tai, C.H.; Lin, C.H.; Wu, R.M. The impact of nonmotor symptoms on quality of life in patients with Parkinson’s disease in Taiwan. Neuropsychiatr. Dis. Treat. 2015, 11, 2865–2873. [Google Scholar] [CrossRef] [Green Version]
  76. Bobholz, J.A.; Rao, S.M. Cognitive dysfunction in multiple sclerosis: A review of recent developments. Curr. Opin. Neurol. 2003, 16, 283–288. [Google Scholar] [CrossRef]
  77. Prakash, R.S.; Schirda, B.; Valentine, T.R.; Crotty, M.; Nicholas, J.A. Emotion dysregulation in multiple sclerosis: Impact on symptoms of depression and anxiety. Mult. Scler. Relat. Disord. 2019, 36, 101399. [Google Scholar] [CrossRef]
  78. Ransome, M.I.; Renoir, T.; Hannan, A.J. Hippocampal neurogenesis, cognitive deficits and affective disorder in Huntington’s disease. Neural. Plast. 2012, 2012, 874387. [Google Scholar] [CrossRef]
  79. Connolly, B.; Fox, S.H. Treatment of cognitive, psychiatric, and affective disorders associated with Parkinson’s disease. Neurotherapeutics 2014, 11, 78–91. [Google Scholar] [CrossRef] [Green Version]
  80. Sampath, D.; Sathyanesan, M.; Newton, S.S. Cognitive dysfunction in major depression and Alzheimer’s disease is associated with hippocampal-prefrontal cortex dysconnectivity. Neuropsychiatr. Dis. Treat. 2017, 13, 1509–1519. [Google Scholar] [CrossRef] [Green Version]
  81. McGregor, G.; Harvey, J. Regulation of Hippocampal Synaptic Function by the Metabolic Hormone, Leptin: Implications for Health and Neurodegenerative Disease. Front Cell Neurosci. 2018, 12, 340. [Google Scholar] [CrossRef] [PubMed]
  82. Mufson, E.J.; Mahady, L.; Waters, D.; Counts, S.E.; Perez, S.E.; DeKosky, S.T.; Ginsberg, S.D.; Ikonomovic, M.D.; Scheff, S.W.; Binder, L.I. Hippocampal plasticity during the progression of Alzheimer’s disease. Neuroscience 2015, 309, 51–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Irving, A.; Harvey, J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog. Lipid. Res. 2021, 82, 101098. [Google Scholar] [CrossRef] [PubMed]
  84. Selkoe, D.J. Alzheimer’s disease: Genes, proteins, and therapy. Physiol. Rev. 2001, 81, 741–766. [Google Scholar] [CrossRef]
  85. Reitz, C.; Mayeux, R. Alzheimer disease: Epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 2014, 88, 640–651. [Google Scholar] [CrossRef] [Green Version]
  86. Goedert, M.; Spillantini, M.G. A century of Alzheimer’s disease. Science 2006, 314, 777–781. [Google Scholar] [CrossRef] [Green Version]
  87. Fotuhi, S.N.; Khalaj-Kondori, M.; Feizi, M.A.H.; Talebi, M. Memory-related process in physiological status and alzheimer’s disease. Mol. Biol. Rep. 2020, 47, 4651–4657. [Google Scholar] [CrossRef]
  88. Brun, A.; Englund, E. Regional pattern of degeneration in Alzheimer’s disease: Neuronal loss and histopathological grading. Histopathology 1981, 5, 549–564. [Google Scholar] [CrossRef]
  89. Wright, A.L.; Zinn, R.; Hohensinn, B.; Konen, L.M.; Beynon, S.B.; Tan, R.P.; Clark, I.A.; Abdipranoto, A.; Vissel, B. Neuroinflammation and neuronal loss precede Abeta plaque deposition in the hAPP-J20 mouse model of Alzheimer’s disease. PLoS ONE 2013, 8, e59586. [Google Scholar] [CrossRef] [Green Version]
  90. Scheff, S.W.; Price, D.A. Alzheimer’s disease-related alterations in synaptic density: Neocortex and hippocampus. J. Alzheimers Dis. 2006, 9, 101–115. [Google Scholar] [CrossRef]
  91. Jahn, H. Memory loss in Alzheimer’s disease. Dialogues Clin. Neurosci. 2013, 15, 445–454. [Google Scholar] [CrossRef] [PubMed]
  92. de Flores, R.; La Joie, R.; Chetelat, G. Structural imaging of hippocampal subfields in healthy aging and Alzheimer’s disease. Neuroscience 2015, 309, 29–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Schönheit, B.; Zarski, R.; Ohm, T.G. Spatial and temporal relationships between plaques and tangles in Alzheimer-pathology. Neurobiol. Aging 2004, 25, 697–711. [Google Scholar] [CrossRef]
  94. Lace, G.; Savva, G.M.; Forster, G.; de Silva, R.; Brayne, C.; Matthews, F.E.; Barclay, J.J.; Dakin, L.; Ince, P.G.; Wharton, S.B.; et al. Hippocampal tau pathology is related to neuroanatomical connections: An ageing population-based study. Brain 2009, 132, 1324–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kril, J.J.; Patel, S.; Harding, A.J.; Halliday, G.M. Patients with vascular dementia due to microvascular pathology have significant hippocampal neuronal loss. J. Neurol. Neurosurg Psychiatry 2002, 72, 747–751. [Google Scholar] [CrossRef] [Green Version]
  96. Padurariu, M.; Ciobica, A.; Mavroudis, I.; Fotiou, D.; Baloyannis, S. Hippocampal neuronal loss in the CA1 and CA3 areas of Alzheimer’s disease patients. Psychiatr. Danub. 2012, 24, 152–158. [Google Scholar]
  97. Price, J.L.; Ko, A.I.; Wade, M.J.; Tsou, S.K.; McKeel, D.W.; Morris, J.C. Neuron number in the entorhinal cortex and CA1 in preclinical Alzheimer disease. Arch Neurol. 2001, 58, 1395–1402. [Google Scholar] [CrossRef]
  98. Skaper, S.D.; Facci, L.; Zusso, M.; Giusti, P. Synaptic Plasticity, Dementia and Alzheimer Disease. CNS Neurol. Disord. Drug Targets 2017, 16, 220–233. [Google Scholar] [CrossRef]
  99. Scheff, S.W.; Price, D.A. Synaptic density in the inner molecular layer of the hippocampal dentate gyrus in Alzheimer disease. J. Neuropathol. Exp. Neurol. 1998, 57, 1146–1153. [Google Scholar] [CrossRef] [Green Version]
  100. Volpicelli-Daley, L.A.; Luk, K.C.; Patel, T.P.; Tanik, S.A.; Riddle, D.M.; Stieber, A.; Meaney, D.F.; Trojanowski, J.Q.; Lee, V.M. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011, 72, 57–71. [Google Scholar] [CrossRef] [Green Version]
  101. Talantova, M.; Sanz-Blasco, S.; Zhang, X.; Xia, P.; Akhtar, M.W.; Okamoto, S.; Dziewczapolski, G.; Nakamura, T.; Cao, G.; Pratt, A.E.; et al. Abeta induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss. Proc. Natl. Acad. Sci. USA 2013, 110, E2518–E2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yao, P.J.; Zhu, M.; Pyun, E.I.; Brooks, A.I.; Therianos, S.; Meyers, V.E.; Coleman, P.D. Defects in expression of genes related to synaptic vesicle traffickingin frontal cortex of Alzheimer’s disease. Neurobiol. Dis. 2003, 12, 97–109. [Google Scholar] [CrossRef]
  103. Counts, S.E.; Alldred, M.J.; Che, S.; Ginsberg, S.D.; Mufson, E.J. Synaptic gene dysregulation within hippocampal CA1 pyramidal neurons in mild cognitive impairment. Neuropharmacology 2014, 79, 172–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Cuestas Torres, D.M.; Cardenas, F.P. Synaptic plasticity in Alzheimer’s disease and healthy aging. Rev. Neurosci. 2020, 31, 245–268. [Google Scholar] [CrossRef]
  105. Soria Lopez, J.A.; Gonzalez, H.M.; Leger, G.C. Alzheimer’s disease. Handb. Clin. Neurol. 2019, 167, 231–255. [Google Scholar] [CrossRef]
  106. Scheff, S.W.; Price, D.A.; Schmitt, F.A.; Mufson, E.J. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol. Aging 2006, 27, 1372–1384. [Google Scholar] [CrossRef]
  107. Scheff, S.W.; Price, D.A.; Schmitt, F.A.; DeKosky, S.T.; Mufson, E.J. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology 2007, 68, 1501–1508. [Google Scholar] [CrossRef]
  108. Fol, R.; Braudeau, J.; Ludewig, S.; Abel, T.; Weyer, S.W.; Roederer, J.P.; Brod, F.; Audrain, M.; Bemelmans, A.P.; Buchholz, C.J.; et al. Viral gene transfer of APPsalpha rescues synaptic failure in an Alzheimer’s disease mouse model. Acta Neuropathol. 2016, 131, 247–266. [Google Scholar] [CrossRef]
  109. Zhang, W.; Gu, G.J.; Zhang, Q.; Liu, J.H.; Zhang, B.; Guo, Y.; Wang, M.Y.; Gong, Q.Y.; Xu, J.R. NSCs promote hippocampal neurogenesis, metabolic changes and synaptogenesis in APP/PS1 transgenic mice. Hippocampus 2017, 27, 1250–1263. [Google Scholar] [CrossRef]
  110. Peretti, D.; Bastide, A.; Radford, H.; Verity, N.; Molloy, C.; Martin, M.G.; Moreno, J.A.; Steinert, J.R.; Smith, T.; Dinsdale, D.; et al. RBM3 mediates structural plasticity and protective effects of cooling in neurodegeneration. Nature 2015, 518, 236–239. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, R.; Zhang, J.; Wu, Y.; Wang, D.; Feng, G.; Tang, Y.P.; Teng, Z.; Chen, C. Monoacylglycerol lipase is a therapeutic target for Alzheimer’s disease. Cell Rep. 2012, 2, 1329–1339. [Google Scholar] [CrossRef] [Green Version]
  112. Steele, J.W.; Brautigam, H.; Short, J.A.; Sowa, A.; Shi, M.; Yadav, A.; Weaver, C.M.; Westaway, D.; Fraser, P.E.; St George-Hyslop, P.H.; et al. Early fear memory defects are associated with altered synaptic plasticity and molecular architecture in the TgCRND8 Alzheimer’s disease mouse model. J. Comp. Neurol. 2014, 522, 2319–2335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tsai, S.T.; Chen, S.Y.; Lin, S.Z.; Tseng, G.F. Rostral intralaminar thalamic deep brain stimulation ameliorates memory deficits and dendritic regression in beta-amyloid-infused rats. Brain Struct Funct. 2020, 225, 751–761. [Google Scholar] [CrossRef]
  114. Liu, Y.; Bian, H.; Xu, S.; Shu, S.; Jia, J.; Chen, J.; Cao, X.; Bao, X.; Gu, Y.; Xia, S.; et al. Muscone Ameliorates Synaptic Dysfunction and Cognitive Deficits in APP/PS1 Mice. J. Alzheimers Dis. 2020, 76, 491–504. [Google Scholar] [CrossRef] [PubMed]
  115. Hou, J.; Wang, C.; Zhang, M.; Ren, M.; Yang, G.; Qu, Z.; Hu, Y. Safflower Yellow Improves the Synaptic Structural Plasticity by Ameliorating the Disorder of Glutamate Circulation in Abeta1-42-induced AD Model Rats. Neurochem. Res. 2020, 45, 1870–1887. [Google Scholar] [CrossRef] [PubMed]
  116. Jacobsen, J.S.; Wu, C.C.; Redwine, J.M.; Comery, T.A.; Arias, R.; Bowlby, M.; Martone, R.; Morrison, J.H.; Pangalos, M.N.; Reinhart, P.H.; et al. Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5161–5166. [Google Scholar] [CrossRef] [Green Version]
  117. 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-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med. 2008, 14, 837–842. [Google Scholar] [CrossRef] [Green Version]
  118. Inestrosa, N.C.; Varela-Nallar, L. Wnt signaling in the nervous system and in Alzheimer’s disease. J. Mol. Cell Biol. 2014, 6, 64–74. [Google Scholar] [CrossRef] [PubMed]
  119. Sun, L.N.; Qi, J.S.; Gao, R. Physical exercise reserved amyloid-beta induced brain dysfunctions by regulating hippocampal neurogenesis and inflammatory response via MAPK signaling. Brain Res. 2018, 1697, 1–9. [Google Scholar] [CrossRef] [PubMed]
  120. Selkoe, D.J. Alzheimer’s disease is a synaptic failure. Science 2002, 298, 789–791. [Google Scholar] [CrossRef] [Green Version]
  121. Mucke, L.; Masliah, E.; Yu, G.-Q.; Mallory, M.; Rockenstein, E.M.; Tatsuno, G.; Hu, K.; Kholodenko, D.; Johnson-Wood, K.; McConlogue, L. High-level neuronal expression of Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: Synaptotoxicity without plaque formation. J. Neurosci. 2000, 20, 4050–4058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Oddo, S.; Caccamo, A.; Shepherd, J.D.; Murphy, M.P.; Golde, T.E.; Kayed, R.; Metherate, R.; Mattson, M.P.; Akbari, Y.; LaFerla, F.M. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: Intracellular Abeta and synaptic dysfunction. Neuron 2003, 39, 409–421. [Google Scholar] [CrossRef] [Green Version]
  123. Kamenetz, F.; Tomita, T.; Hsieh, H.; Seabrook, G.; Borchelt, D.; Iwatsubo, T.; Sisodia, S.; Malinow, R. APP processing and synaptic function. Neuron 2003, 37, 925–937. [Google Scholar] [CrossRef] [Green Version]
  124. Lang, A.E.; Lozano, A.M. Parkinson’s disease. Second of two parts. N. Engl. J. Med. 1998, 339, 1130–1143. [Google Scholar] [CrossRef] [PubMed]
  125. Dorsey, E.R.; Elbaz, A.; Nichols, E.; Abd-Allah, F.; Abdelalim, A.; Adsuar, J.C.; Ansha, M.G.; Brayne, C.; Choi, J.Y.J.; Collado-Mateo, D.; et al. Global, regional, and national burden of Parkinson’s disease, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018, 17, 939–953. [Google Scholar] [CrossRef] [Green Version]
  126. Collaborators, G. GBD 2016 Dementia Collaborators Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar]
  127. Leroi, I.; McDonald, K.; Pantula, H.; Harbishettar, V. Cognitive impairment in Parkinson disease: Impact on quality of life, disability, and caregiver burden. J. Geriatr. Psychiatry Neurol. 2012, 25, 208–214. [Google Scholar] [CrossRef]
  128. Barone, P.; Erro, R.; Picillo, M. Quality of Life and Nonmotor Symptoms in Parkinson’s Disease. Int. Rev. Neurobiol. 2017, 133, 499–516. [Google Scholar] [CrossRef]
  129. Calabresi, P.; Castrioto, A.; Di Filippo, M.; Picconi, B. New experimental and clinical links between the hippocampus and the dopaminergic system in Parkinson’s disease. Lancet Neurol. 2013, 12, 811–821. [Google Scholar] [CrossRef]
  130. Regensburger, M.; Prots, I.; Winner, B. Adult hippocampal neurogenesis in Parkinson’s disease: Impact on neuronal survival and plasticity. Neural. Plast. 2014, 2014, 454696. [Google Scholar] [CrossRef] [Green Version]
  131. Barone, P.; Antonini, A.; Colosimo, C.; Marconi, R.; Morgante, L.; Avarello, T.P.; Bottacchi, E.; Cannas, A.; Ceravolo, G.; Ceravolo, R.; et al. The PRIAMO study: A multicenter assessment of nonmotor symptoms and their impact on quality of life in Parkinson’s disease. Mov. Disord. 2009, 24, 1641–1649. [Google Scholar] [CrossRef] [PubMed]
  132. Leentjens, A.F.; Van den Akker, M.; Metsemakers, J.F.; Lousberg, R.; Verhey, F.R. Higher incidence of depression preceding the onset of Parkinson’s disease: A register study. Mov. Disord. 2003, 18, 414–418. [Google Scholar] [CrossRef] [PubMed]
  133. Ballanger, B.; Klinger, H.; Eche, J.; Lerond, J.; Vallet, A.E.; Le Bars, D.; Tremblay, L.; Sgambato-Faure, V.; Broussolle, E.; Thobois, S. Role of serotonergic 1A receptor dysfunction in depression associated with Parkinson’s disease. Mov. Disord. 2012, 27, 84–89. [Google Scholar] [CrossRef] [PubMed]
  134. Pavese, N.; Metta, V.; Bose, S.K.; Chaudhuri, K.R.; Brooks, D.J. Fatigue in Parkinson’s disease is linked to striatal and limbic serotonergic dysfunction. Brain 2010, 133, 3434–3443. [Google Scholar] [CrossRef]
  135. Goto, Y.; Grace, A.A. Dopaminergic modulation of limbic and cortical drive of nucleus accumbens in goal-directed behavior. Nat. Neurosci. 2005, 8, 805–812. [Google Scholar] [CrossRef]
  136. Costa, C.; Sgobio, C.; Siliquini, S.; Tozzi, A.; Tantucci, M.; Ghiglieri, V.; Di Filippo, M.; Pendolino, V.; de Iure, A.; Marti, M.; et al. Mechanisms underlying the impairment of hippocampal long-term potentiation and memory in experimental Parkinson’s disease. Brain 2012, 135, 1884–1899. [Google Scholar] [CrossRef] [Green Version]
  137. Esmaeili-Mahani, S.; Haghparast, E.; Nezhadi, A.; Abbasnejad, M.; Sheibani, V. Apelin-13 prevents hippocampal synaptic plasticity impairment in Parkinsonism rats. J. Chem. Neuroanat. 2021, 111, 101884. [Google Scholar] [CrossRef]
  138. Pendolino, V.; Bagetta, V.; Ghiglieri, V.; Sgobio, C.; Morelli, E.; Poggini, S.; Branchi, I.; Latagliata, E.C.; Pascucci, T.; Puglisi-Allegra, S.; et al. l-DOPA reverses the impairment of Dentate Gyrus LTD in experimental parkinsonism via beta-adrenergic receptors. Exp. Neurol. 2014, 261, 377–385. [Google Scholar] [CrossRef]
  139. Wang, Y.; Feng, L.; Liu, S.; Zhou, X.; Yin, T.; Liu, Z.; Yang, Z. Transcranial magneto-acoustic stimulation improves neuroplasticity in hippocampus of Parkinson’s disease model mice. Neurotherapeutics 2019, 16, 1210–1224. [Google Scholar] [CrossRef] [Green Version]
  140. Wu, Q.; Takano, H.; Riddle, D.M.; Trojanowski, J.Q.; Coulter, D.A.; Lee, V.M. alpha-Synuclein (alphaSyn) Preformed Fibrils Induce Endogenous alphaSyn Aggregation, Compromise Synaptic Activity and Enhance Synapse Loss in Cultured Excitatory Hippocampal Neurons. J. Neurosci. 2019, 39, 5080–5094. [Google Scholar] [CrossRef] [Green Version]
  141. Swanson-Park, J.; Coussens, C.; Mason-Parker, S.; Raymond, C.; Hargreaves, E.; Dragunow, M.; Cohen, A.; Abraham, W. A double dissociation within the hippocampus of dopamine D1/D5 receptor and β-adrenergic receptor contributions to the persistence of long-term potentiation. Neuroscience 1999, 92, 485–497. [Google Scholar] [CrossRef]
  142. Hansen, N.; Manahan-Vaughan, D. Dopamine D1/D5 receptors mediate informational saliency that promotes persistent hippocampal long-term plasticity. Cereb. Cortex. 2014, 24, 845–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kusuki, T.; Imahori, Y.; Ueda, S.; Inokuchi, K. Dopaminergic modulation of LTP induction in the dentate gyrus of intact brain. Neuroreport 1997, 8, 2037–2040. [Google Scholar] [CrossRef] [PubMed]
  144. O’Carroll, C.M.; Martin, S.J.; Sandin, J.; Frenguelli, B.; Morris, R.G. Dopaminergic modulation of the persistence of one-trial hippocampus-dependent memory. Learn Mem. 2006, 13, 760–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Frey, U.; Morris, R.G. Synaptic tagging and long-term potentiation. Nature 1997, 385, 533–536. [Google Scholar] [CrossRef]
  146. Calabresi, P.; Ghiglieri, V.; Mazzocchetti, P.; Corbelli, I.; Picconi, B. Levodopa-induced plasticity: A double-edged sword in Parkinson’s disease? Philos. Trans. R. Soc. Lond. B Biol. Sci. 2015, 370, 20140184. [Google Scholar] [CrossRef] [Green Version]
  147. Borodovitsyna, O.; Flamini, M.; Chandler, D. Noradrenergic Modulation of Cognition in Health and Disease. Neural. Plast. 2017, 2017, 6031478. [Google Scholar] [CrossRef] [Green Version]
  148. Kohl, Z.; Ben Abdallah, N.; Vogelgsang, J.; Tischer, L.; Deusser, J.; Amato, D.; Anderson, S.; Muller, C.P.; Riess, O.; Masliah, E.; et al. Severely impaired hippocampal neurogenesis associates with an early serotonergic deficit in a BAC alpha-synuclein transgenic rat model of Parkinson’s disease. Neurobiol. Dis. 2016, 85, 206–217. [Google Scholar] [CrossRef] [Green Version]
  149. Cubo, E.; Shannon, K.M.; Tracy, D.; Jaglin, J.A.; Bernard, B.A.; Wuu, J.; Leurgans, S.E. Effect of donepezil on motor and cognitive function in Huntington disease. Neurology 2006, 67, 1268–1271. [Google Scholar] [CrossRef]
  150. Paulsen, J.S. Cognitive impairment in Huntington disease: Diagnosis and treatment. Curr. Neurol. Neurosci. Rep. 2011, 11, 474–483. [Google Scholar] [CrossRef] [Green Version]
  151. Craufurd, D.; Thompson, J.C.; Snowden, J.S. Behavioral changes in Huntington Disease. Neuropsychiatry Neuropsychol. Behav. Neurol. 2001, 14, 219–226. [Google Scholar]
  152. Bates, G.; Tabrizi, S.; Jones, L. Huntington’s Disease; Oxford Monographs on Medical G; Oxford University Press: New York, NY, USA, 2014. [Google Scholar]
  153. The Huntington’s Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 1993, 72, 971–983. [Google Scholar] [CrossRef]
  154. Waldvogel, H.J.; Kim, E.H.; Tippett, L.J.; Vonsattel, J.-P.G.; Faull, R.L. The Neuropathology of Huntington’s Disease. Curr. Top Behav. Neurosci. 2015, 22, 33–80. [Google Scholar] [CrossRef] [PubMed]
  155. Rubinstein, D. Molecular biology of Huntingtons Disease (HD) and HD-like disorders. In Genetics of Movement Disorders; Pulst, S., Ed.; Academic Press: Cambridge, MA, USA, 2003; pp. 365–377. [Google Scholar]
  156. Vonsattel, J.P.; DiFiglia, M. Huntington disease. J. Neuropathol. Exp. Neurol. 1998, 57, 369–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Shehadeh, J.; Fernandes, H.B.; Zeron Mullins, M.M.; Graham, R.K.; Leavitt, B.R.; Hayden, M.R.; Raymond, L.A. Striatal neuronal apoptosis is preferentially enhanced by NMDA receptor activation in YAC transgenic mouse model of Huntington disease. Neurobiol. Dis. 2006, 21, 392–403. [Google Scholar] [CrossRef] [PubMed]
  158. Rikani, A.A.; Choudhry, Z.; Choudhry, A.M.; Rizvi, N.; Ikram, H.; Mobassarah, N.J.; Tulli, S. The mechanism of degeneration of striatal neuronal subtypes in Huntington disease. Ann. Neurosci. 2014, 21, 112–114. [Google Scholar] [CrossRef]
  159. Lebouc, M.; Richard, Q.; Garret, M.; Baufreton, J. Striatal circuit development and its alterations in Huntington’s disease. Neurobiol. Dis. 2020, 145, 105076. [Google Scholar] [CrossRef]
  160. Spargo, E.; Everall, I.P.; Lantos, P.L. Neuronal loss in the hippocampus in Huntington’s disease: A comparison with HIV infection. J. Neurol. Neurosurg. Psychiatry 1993, 56, 487–491. [Google Scholar] [CrossRef]
  161. Cheong, R.Y.; Gabery, S.; Petersen, A. The Role of Hypothalamic Pathology for Non-Motor Features of Huntington’s Disease. J. Huntingt. Dis. 2019, 8, 375–391. [Google Scholar] [CrossRef] [Green Version]
  162. Paulsen, J.S.; Langbehn, D.R.; Stout, J.C.; Aylward, E.; Ross, C.A.; Nance, M.; Guttman, M.; Johnson, S.; MacDonald, M.; Beglinger, L.J.; et al. Detection of Huntington’s disease decades before diagnosis: The Predict-HD study. J. Neurol. Neurosurg. Psychiatry. 2008, 79, 874–880. [Google Scholar] [CrossRef] [Green Version]
  163. Wilkie, C.M.; Barnes, J.R.; Benson, C.M.; Brymer, K.J.; Nafar, F.; Parsons, M.P. Hippocampal Synaptic Dysfunction in a Mouse Model of Huntington Disease Is Not Alleviated by Ceftriaxone Treatment. eNeuro 2020, 7, 19. [Google Scholar] [CrossRef] [PubMed]
  164. Usdin, M.T.; Shelbourne, P.F.; Myers, R.M.; Madison, D.V. Impaired synaptic plasticity in mice carrying the Huntington’s disease mutation. Hum. Mol. Genet. 1999, 8, 839–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Murphy, K.P.; Carter, R.J.; Lione, L.A.; Mangiarini, L.; Mahal, A.; Bates, G.P.; Dunnett, S.B.; Morton, A.J. Abnormal synaptic plasticity and impaired spatial cognition in mice transgenic for exon 1 of the human Huntington’s disease mutation. J. Neurosci. 2000, 20, 5115–5123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Cummings, D.M.; Milnerwood, A.J.; Dallerac, G.M.; Waights, V.; Brown, J.Y.; Vatsavayai, S.C.; Hirst, M.C.; Murphy, K.P. Aberrant cortical synaptic plasticity and dopaminergic dysfunction in a mouse model of Huntington’s disease. Hum. Mol. Genet. 2006, 15, 2856–2868. [Google Scholar] [CrossRef] [Green Version]
  167. Milnerwood, A.J.; Cummings, D.M.; Dallerac, G.M.; Brown, J.Y.; Vatsavayai, S.C.; Hirst, M.C.; Rezaie, P.; Murphy, K.P. Early development of aberrant synaptic plasticity in a mouse model of Huntington’s disease. Hum. Mol. Genet. 2006, 15, 1690–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Lynch, G.; Kramar, E.A.; Rex, C.S.; Jia, Y.; Chappas, D.; Gall, C.M.; Simmons, D.A. Brain-derived neurotrophic factor restores synaptic plasticity in a knock-in mouse model of Huntington’s disease. J. Neurosci. 2007, 27, 4424–4434. [Google Scholar] [CrossRef]
  169. Simmons, D.A.; Rex, C.S.; Palmer, L.; Pandyarajan, V.; Fedulov, V.; Gall, C.M.; Lynch, G. Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington’s disease knockin mice. Proc. Natl. Acad. Sci. USA 2009, 106, 4906–4911. [Google Scholar] [CrossRef] [Green Version]
  170. Brito, V.; Giralt, A.; Enriquez-Barreto, L.; Puigdellivol, M.; Suelves, N.; Zamora-Moratalla, A.; Ballesteros, J.J.; Martin, E.D.; Dominguez-Iturza, N.; Morales, M.; et al. Neurotrophin receptor p75(NTR) mediates Huntington’s disease-associated synaptic and memory dysfunction. J. Clin. Investig. 2014, 124, 4411–4428. [Google Scholar] [CrossRef] [Green Version]
  171. Kolodziejczyk, K.; Parsons, M.P.; Southwell, A.L.; Hayden, M.R.; Raymond, L.A. Striatal synaptic dysfunction and hippocampal plasticity deficits in the Hu97/18 mouse model of Huntington disease. PLoS ONE 2014, 9, e94562. [Google Scholar] [CrossRef]
  172. Giralt, A.; Brito, V.; Chevy, Q.; Simonnet, C.; Otsu, Y.; Cifuentes-Diaz, C.; de Pins, B.; Coura, R.; Alberch, J.; Gines, S.; et al. Pyk2 modulates hippocampal excitatory synapses and contributes to cognitive deficits in a Huntington’s disease model. Nat. Commun. 2017, 8, 15592. [Google Scholar] [CrossRef]
  173. Sepers, M.D.; Smith-Dijak, A.; LeDue, J.; Kolodziejczyk, K.; Mackie, K.; Raymond, L.A. Endocannabinoid-Specific Impairment in Synaptic Plasticity in Striatum of Huntington’s Disease Mouse Model. J. Neurosci. 2018, 38, 544–554. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, H.; Zhang, C.; Vincent, J.; Zala, D.; Benstaali, C.; Sainlos, M.; Grillo-Bosch, D.; Daburon, S.; Coussen, F.; Cho, Y.; et al. Modulation of AMPA receptor surface diffusion restores hippocampal plasticity and memory in Huntington’s disease models. Nat. Commun. 2018, 9, 4272. [Google Scholar] [CrossRef] [PubMed]
  175. Hodgson, J.G.; Agopyan, N.; Gutekunst, C.A.; Leavitt, B.R.; LePiane, F.; Singaraja, R.; Smith, D.J.; Bissada, N.; McCutcheon, K.; Nasir, J.; et al. A YAC mouse model for Huntington’s disease with full-length mutant huntingtin, cytoplasmic toxicity, and selective striatal neurodegeneration. Neuron 1999, 23, 181–192. [Google Scholar] [CrossRef] [Green Version]
  176. Quirion, J.G.; Parsons, M.P. The Onset and Progression of Hippocampal Synaptic Plasticity Deficits in the Q175FDN Mouse Model of Huntington Disease. Front. Cell Neurosci. 2019, 13, 326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Anglada-Huguet, M.; Xifro, X.; Giralt, A.; Zamora-Moratalla, A.; Martin, E.D.; Alberch, J. Prostaglandin E2 EP1 receptor antagonist improves motor deficits and rescues memory decline in R6/1 mouse model of Huntington’s disease. Mol. Neurobiol. 2014, 49, 784–795. [Google Scholar] [CrossRef] [PubMed]
  178. Bruel-Jungerman, E.; Davis, S.; Laroche, S. Brain plasticity mechanisms and memory: A party of four. Neuroscientist 2007, 13, 492–505. [Google Scholar] [CrossRef] [PubMed]
  179. Bruel-Jungerman, E.; Rampon, C.; Laroche, S. Adult hippocampal neurogenesis, synaptic plasticity and memory: Facts and hypotheses. Rev. Neurosci. 2007, 18, 93–114. [Google Scholar] [CrossRef]
  180. Di Filippo, M.; Tozzi, A.; Picconi, B.; Ghiglieri, V.; Calabresi, P. Plastic abnormalities in experimental Huntington’s disease. Curr. Opin. Pharmacol. 2007, 7, 106–111. [Google Scholar] [CrossRef]
  181. Cattaneo, E.; Zuccato, C.; Tartari, M. Normal huntingtin function: An alternative approach to Huntington’s disease. Nat. Rev. Neurosci. 2005, 6, 919–930. [Google Scholar] [CrossRef]
  182. Cotsapas, C.; Mitrovic, M.; Hafler, D. Multiple sclerosis. Handb. Clin. Neurol. 2018, 148, 723–730. [Google Scholar] [CrossRef]
  183. Karussis, D. The diagnosis of multiple sclerosis and the various related demyelinating syndromes: A critical review. J. Autoimmun. 2014, 48–49, 134–142. [Google Scholar] [CrossRef]
  184. Trapp, B.D.; Peterson, J.; Ransohoff, R.M.; Rudick, R.; Mork, S.; Bo, L. Axonal transection in the lesions of multiple sclerosis. N. Engl. J. Med. 1998, 338, 278–285. [Google Scholar] [CrossRef] [PubMed]
  185. Lassmann, H. Pathology and disease mechanisms in different stages of multiple sclerosis. J. Neurol. Sci. 2013, 333, 1–4. [Google Scholar] [CrossRef] [PubMed]
  186. Vercellino, M.; Plano, F.; Votta, B.; Mutani, R.; Giordana, M.T.; Cavalla, P. Grey matter pathology in multiple sclerosis. J. Neuropathol. Exp. Neurol. 2005, 64, 1101–1107. [Google Scholar] [CrossRef] [PubMed]
  187. Geurts, J.J.; Bo, L.; Roosendaal, S.D.; Hazes, T.; Daniels, R.; Barkhof, F.; Witter, M.P.; Huitinga, I.; van der Valk, P. Extensive hippocampal demyelination in multiple sclerosis. J. Neuropathol. Exp. Neurol. 2007, 66, 819–827. [Google Scholar] [CrossRef] [Green Version]
  188. Dutta, R.; Chang, A.; Doud, M.K.; Kidd, G.J.; Ribaudo, M.V.; Young, E.A.; Fox, R.J.; Staugaitis, S.M.; Trapp, B.D. Demyelination causes synaptic alterations in hippocampi from multiple sclerosis patients. Ann. Neurol. 2011, 69, 445–454. [Google Scholar] [CrossRef]
  189. Drake, M.A.; Carra, A.; Allegri, R.F.; Luetic, G. Differential patterns of memory performance in relapsing, remitting and secondary progressive multiple sclerosis. Neurol. India 2006, 54, 370–376. [Google Scholar] [CrossRef]
  190. Chiaravalloti, N.D.; DeLuca, J. Cognitive impairment in multiple sclerosis. Lancet Neurol. 2008, 7, 1139–1151. [Google Scholar] [CrossRef]
  191. Gaudino, E.A.; Chiaravalloti, N.D.; DeLuca, J.; Diamond, B.J. A comparison of memory performance in relapsing-remitting, primary progressive and secondary progressive, multiple sclerosis. Neuropsychiatry Neuropsychol. Behav. Neurol. 2001, 14, 32–44. [Google Scholar]
  192. Beatty, W.W.; Wilbanks, S.L.; Blanco, C.R.; Hames, K.A.; Tivis, R.; Paul, R.H. Memory disturbance in multiple sclerosis: Reconsideration of patterns of performance on the selective reminding test. J. Clin. Exp. Neuropsychol. 1996, 18, 56–62. [Google Scholar] [CrossRef]
  193. Litvan, I.; Grafman, J.; Vendrell, P.; Martinez, J.M. Slowed information processing in multiple sclerosis. Arch. Neurol. 1988, 45, 281–285. [Google Scholar] [CrossRef] [PubMed]
  194. Feinstein, A.; Magalhaes, S.; Richard, J.F.; Audet, B.; Moore, C. The link between multiple sclerosis and depression. Nat. Rev. Neurol. 2014, 10, 507–517. [Google Scholar] [CrossRef] [PubMed]
  195. Rocca, M.A.; Barkhof, F.; De Luca, J.; Frisen, J.; Geurts, J.J.G.; Hulst, H.E.; Sastre-Garriga, J.; Filippi, M.; Group, M.S. The hippocampus in multiple sclerosis. Lancet Neurol. 2018, 17, 918–926. [Google Scholar] [CrossRef]
  196. Pelletier, J.; Audoin, B.; Reuter, F.; Ranjeva, J. Plasticity in MS: From Functional Imaging to Rehabilitation. Int. MS J. 2009, 16, 26–31. [Google Scholar]
  197. Amato, M.P.; Zipoli, V.; Portaccio, E. Cognitive changes in multiple sclerosis. Expert Rev. Neurother. 2008, 8, 1585–1596. [Google Scholar] [CrossRef]
  198. Nistico, R.; Mori, F.; Feligioni, M.; Nicoletti, F.; Centonze, D. Synaptic plasticity in multiple sclerosis and in experimental autoimmune encephalomyelitis. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130162. [Google Scholar] [CrossRef] [Green Version]
  199. Michailidou, I.; Willems, J.G.; Kooi, E.J.; van Eden, C.; Gold, S.M.; Geurts, J.J.; Baas, F.; Huitinga, I.; Ramaglia, V. Complement C 1q-C 3–associated synaptic changes in multiple sclerosis hippocampus. Ann. Neurol. 2015, 77, 1007–1026. [Google Scholar] [CrossRef]
  200. Di Filippo, M.; Chiasserini, D.; Gardoni, F.; Viviani, B.; Tozzi, A.; Giampà, C.; Costa, C.; Tantucci, M.; Zianni, E.; Boraso, M.; et al. Effects of central and peripheral inflammation on hippocampal synaptic plasticity. Neurobiol. Dis. 2013, 52, 229–236. [Google Scholar] [CrossRef]
  201. Kim, D.Y.; Hao, J.; Liu, R.; Turner, G.; Shi, F.-D.; Rho, J.M. Inflammation-Mediated Memory Dysfunction and Effects of a Ketogenic Diet in a Murine Model of Multiple Sclerosis. PLoS ONE 2012, 7, e35476. [Google Scholar] [CrossRef] [Green Version]
  202. Di Filippo, M.; De Iure, A.; Giampà, C.; Chiasserini, D.; Tozzi, A.; Orvietani, P.L.; Ghiglieri, V.; Tantucci, M.; Durante, V.; Quiroga-Varela, A. Persistent activation of microglia and NADPH oxidase drive hippocampal dysfunction in experimental multiple sclerosis. Sci. Rep. 2016, 6, 1–16. [Google Scholar]
  203. Novkovic, T.; Shchyglo, O.; Gold, R.; Manahan-Vaughan, D. Hippocampal function is compromised in an animal model of multiple sclerosis. Neuroscience 2015, 309, 100–112. [Google Scholar] [CrossRef]
  204. Prochnow, N.; Gold, R.; Haghikia, A. An electrophysiologic approach to quantify impaired synaptic transmission and plasticity in experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2013, 264, 48–53. [Google Scholar] [CrossRef] [PubMed]
  205. Nisticò, R.; Mango, D.; Mandolesi, G.; Piccinin, S.; Berretta, N.; Pignatelli, M.; Feligioni, M.; Musella, A.; Gentile, A.; Mori, F.; et al. Inflammation Subverts Hippocampal Synaptic Plasticity in Experimental Multiple Sclerosis. PLoS ONE 2013, 8, e54666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Di Filippo, M.; de Iure, A.; Durante, V.; Gaetani, L.; Mancini, A.; Sarchielli, P.; Calabresi, P. Synaptic plasticity and experimental autoimmune encephalomyelitis: Implications for multiple sclerosis. Brain Res. 2015, 1621, 205–213. [Google Scholar] [CrossRef] [PubMed]
  207. Mori, F.; Nistico, R.; Mandolesi, G.; Piccinin, S.; Mango, D.; Kusayanagi, H.; Berretta, N.; Bergami, A.; Gentile, A.; Musella, A.; et al. Interleukin-1β Promotes Long-Term Potentiation in Patients with Multiple Sclerosis. NeuroMolecular Med. 2014, 16, 38–51. [Google Scholar] [CrossRef]
  208. Mosayebi, G.; Soleyman, M.R.; Khalili, M.; Mosleh, M.; Palizvan, M.R. Changes in Synaptic Transmission and Long-term Potentiation Induction as a Possible Mechanism for Learning Disability in an Animal Model of Multiple Sclerosis. Int. Neurourol. J. 2016, 20, 26–32. [Google Scholar] [CrossRef] [Green Version]
  209. Viviani, B.; Gardoni, F.; Bartesaghi, S.; Corsini, E.; Facchi, A.; Galli, C.L.; Di Luca, M.; Marinovich, M. Interleukin-1β Released by gp120 Drives Neural Death through Tyrosine Phosphorylation and Trafficking of NMDA Receptors. J. Biol. Chem. 2006, 281, 30212–30222. [Google Scholar] [CrossRef] [Green Version]
  210. Coogan, A.; O’Connor, J.J. Inhibition of NMDA receptor-mediated synaptic transmission in the rat dentate gyrus in vitro by IL-1β. Neuroreport 1997, 8, 2107–2110. [Google Scholar] [CrossRef]
  211. Lai, A.Y.; Swayze, R.D.; El-Husseini, A.; Song, C. Interleukin-1 beta modulates AMPA receptor expression and phosphorylation in hippocampal neurons. J. Neuroimmunol. 2006, 175, 97–106. [Google Scholar] [CrossRef]
  212. Mancini, A.; Gaetani, L.; Di Gregorio, M.; Tozzi, A.; Ghiglieri, V.; Calabresi, P.; Di Filippo, M. Hippocampal neuroplasticity and inflammation: Relevance for multiple sclerosis. Mult. Scler. Demyelinating Disord. 2017, 2, 2. [Google Scholar] [CrossRef] [Green Version]
  213. Luhder, F.; Gold, R.; Flugel, A.; Linker, R.A. Brain-derived neurotrophic factor in neuroimmunology: Lessons learned from multiple sclerosis patients and experimental autoimmune encephalomyelitis models. Arch. Immunol. Ther. Exp. 2013, 61, 95–105. [Google Scholar] [CrossRef] [PubMed]
  214. Frisoni, G.B.; Laakso, M.P.; Beltramello, A.; Geroldi, C.; Bianchetti, A.; Soininen, H.; Trabucchi, M. Hippocampal and entorhinal cortex atrophy in frontotemporal dementia and Alzheimer’s disease. Neurology 1999, 52, 91–100. [Google Scholar] [CrossRef] [PubMed]
  215. Laakso, M.P.; Frisoni, G.B.; Kononen, M.; Mikkonen, M.; Beltramello, A.; Geroldi, C.; Bianchetti, A.; Trabucchi, M.; Soininen, H.; Aronen, H.J. Hippocampus and entorhinal cortex in frontotemporal dementia and Alzheimer’s disease: A morphometric MRI study. Biol. Psychiatry. 2000, 47, 1056–1063. [Google Scholar] [CrossRef]
  216. Graff-Radford, J. Vascular Cognitive Impairment. Contin. Lifelong Learn. Neurol. 2019, 25, 147–164. [Google Scholar] [CrossRef] [PubMed]
  217. Appleton, J.P.; Scutt, P.; Sprigg, N.; Bath, P.M. Hypercholesterolaemia and vascular dementia. Clin. Sci. 2017, 131, 1561–1578. [Google Scholar] [CrossRef] [Green Version]
  218. Bocchetta, M.; Iglesias, J.E.; Scelsi, M.A.; Cash, D.M.; Cardoso, M.J.; Modat, M.; Altmann, A.; Ourselin, S.; Warren, J.D.; Rohrer, J.D. Hippocampal Subfield Volumetry: Differential Pattern of Atrophy in Different Forms of Genetic Frontotemporal Dementia. J. Alzheimers Dis. 2018, 64, 497–504. [Google Scholar] [CrossRef] [Green Version]
  219. Wilson, N.A.; Ramanan, S.; Roquet, D.; Goldberg, Z.L.; Hodges, J.R.; Piguet, O.; Irish, M. Scene construction impairments in frontotemporal dementia: Evidence for a primary hippocampal contribution. Neuropsychologia 2020, 137, 107327. [Google Scholar] [CrossRef]
  220. Christidi, F.; Karavasilis, E.; Velonakis, G.; Ferentinos, P.; Rentzos, M.; Kelekis, N.; Evdokimidis, I.; Bede, P. The Clinical and Radiological Spectrum of Hippocampal Pathology in Amyotrophic Lateral Sclerosis. Front. Neurol. 2018, 9, 523. [Google Scholar] [CrossRef] [Green Version]
  221. Liu, B.; Liu, J.; Zhang, J.; Mao, W.; Li, S. Effects of autophagy on synaptic-plasticity-related protein expression in the hippocampus CA1 of a rat model of vascular dementia. Neurosci. Lett. 2019, 707, 134312. [Google Scholar] [CrossRef]
  222. Huang, Y.; Liao, X.; Wang, H.; Luo, J.; Zhong, S.; Zhang, Z.; Zhang, F.; Chen, J.; Xie, F. Effects of imperatorin on apoptosis and synaptic plasticity in vascular dementia rats. Sci. Rep. 2021, 11, 8590. [Google Scholar] [CrossRef]
  223. Li, X.; Lu, F.; Li, W.; Qin, L.; Yao, Y.; Ge, X.; Yu, Q.; Liang, X.; Zhao, D.; Li, X.; et al. Edaravone injection reverses learning and memory deficits in a rat model of vascular dementia. Acta Biochim. Biophys. Sin. 2017, 49, 83–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Yang, H.Y.; Liu, Y.; Xie, J.C.; Liu, N.N.; Tian, X. Effects of repetitive transcranial magnetic stimulation on synaptic plasticity and apoptosis in vascular dementia rats. Behav. Brain Res. 2015, 281, 149–155. [Google Scholar] [CrossRef] [PubMed]
  225. Meftahi, G.H.; Bayat, M.; Zarifkar, A.H.; Akbari, S.; Haghighi, A.B.; Naseh, M.; Nejad, A.Y.; Haghani, M. Treatment with edaravone improves the structure and functional changes in the hippocampus after chronic cerebral hypoperfusion in rat. Brain Res. Bull. 2021, 174, 122–130. [Google Scholar] [CrossRef]
  226. Levine, D.A.; Langa, K.M. Vascular cognitive impairment: Disease mechanisms and therapeutic implications. Neurotherapeutics 2011, 8, 361–373. [Google Scholar] [CrossRef]
  227. Radzicki, D.; Liu, E.; Deng, H.X.; Siddique, T.; Martina, M. Early Impairment of Synaptic and Intrinsic Excitability in Mice Expressing ALS/Dementia-Linked Mutant UBQLN2. Front Cell Neurosci. 2016, 10, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Ho, W.Y.; Navakkode, S.; Liu, F.; Soong, T.W.; Ling, S.C. Deregulated expression of a longevity gene, Klotho, in the C9orf72 deletion mice with impaired synaptic plasticity and adult hippocampal neurogenesis. Acta Neuropathol. Commun. 2020, 8, 155. [Google Scholar] [CrossRef] [PubMed]
  229. Christidi, F.; Karavasilis, E.; Rentzos, M.; Velonakis, G.; Zouvelou, V.; Xirou, S.; Argyropoulos, G.; Papatriantafyllou, I.; Pantolewn, V.; Ferentinos, P.; et al. Hippocampal pathology in amyotrophic lateral sclerosis: Selective vulnerability of subfields and their associated projections. Neurobiol. Aging 2019, 84, 178–188. [Google Scholar] [CrossRef]
  230. Brettschneider, J.; Del Tredici, K.; Toledo, J.B.; Robinson, J.L.; Irwin, D.J.; Grossman, M.; Suh, E.R.; Van Deerlin, V.M.; Wood, E.M.; Baek, Y.; et al. Stages of pTDP-43 pathology in amyotrophic lateral sclerosis. Ann. Neurol. 2013, 74, 20–38. [Google Scholar] [CrossRef]
  231. Bueno, A.P.A.; Pinaya, W.H.L.; Moura, L.M.; Bertoux, M.; Radakovic, R.; Kiernan, M.C.; Teixeira, A.L.; de Souza, L.C.; Hornberger, M.; Sato, J.R. Structural and functional papez circuit integrity in amyotrophic lateral sclerosis. Brain Imaging Behav. 2018, 12, 1622–1630. [Google Scholar] [CrossRef] [Green Version]
  232. Christidi, F.; Karavasilis, E.; Zalonis, I.; Ferentinos, P.; Giavri, Z.; Wilde, E.A.; Xirou, S.; Rentzos, M.; Zouvelou, V.; Velonakis, G.; et al. Memory-related white matter tract integrity in amyotrophic lateral sclerosis: An advanced neuroimaging and neuropsychological study. Neurobiol. Aging 2017, 49, 69–78. [Google Scholar] [CrossRef]
  233. Trojsi, F.; Di Nardo, F.; Caiazzo, G.; Siciliano, M.; D’Alvano, G.; Ferrantino, T.; Passaniti, C.; Ricciardi, D.; Esposito, S.; Lavorgna, L.; et al. Hippocampal connectivity in Amyotrophic Lateral Sclerosis (ALS): More than Papez circuit impairment. Brain Imaging Behav. 2021, 15, 2126–2138. [Google Scholar] [CrossRef] [PubMed]
  234. Rei, N.; Rombo, D.M.; Ferreira, M.F.; Baqi, Y.; Muller, C.E.; Ribeiro, J.A.; Sebastiao, A.M.; Vaz, S.H. Hippocampal synaptic dysfunction in the SOD1(G93A) mouse model of Amyotrophic Lateral Sclerosis: Reversal by adenosine A2AR blockade. Neuropharmacology 2020, 171, 108106. [Google Scholar] [CrossRef] [PubMed]
  235. Spalloni, A.; Geracitano, R.; Berretta, N.; Sgobio, C.; Bernardi, G.; Mercuri, N.B.; Longone, P.; Ammassari-Teule, M. Molecular and synaptic changes in the hippocampus underlying superior spatial abilities in pre-symptomatic G93A+/+ mice overexpressing the human Cu/Zn superoxide dismutase (Gly93→ ALA) mutation. Exp. Neurol. 2006, 197, 505–514. [Google Scholar] [CrossRef] [PubMed]
  236. Zhao, X.; Liu, J.; Yang, S.; Song, D.; Wang, C.; Chen, C.; Li, X.; Wang, Q.; Ge, S.; Yang, R.; et al. Ling-Yang-Gou-Teng-decoction prevents vascular dementia through inhibiting oxidative stress induced neurovascular coupling dysfunction. J. Ethnopharmacol. 2018, 222, 229–238. [Google Scholar] [CrossRef] [PubMed]
  237. Liu, B.; Tang, J.; Li, S.; Zhang, J.; Yuan, M.; Wang, R. Autophagy activation aggravates neuronal injury in the hippocampus of vascular dementia rats. Neural Regen. Res. 2014, 9, 1288–1296. [Google Scholar] [CrossRef]
  238. He, Z.; Hu, M.; Zha, Y.H.; Li, Z.C.; Zhao, B.; Yu, L.L.; Yu, M.; Qian, Y. Piracetam ameliorated oxygen and glucose deprivation-induced injury in rat cortical neurons via inhibition of oxidative stress, excitatory amino acids release and P53/Bax. Cell Mol. Neurobiol. 2014, 34, 539–547. [Google Scholar] [CrossRef]
  239. Dong, J.; Zhao, J.; Lin, Y.; Liang, H.; He, X.; Zheng, X.; Sui, M.; Zhuang, Z.; Yan, T. Exercise improves recognition memory and synaptic plasticity in the prefrontal cortex for rats modelling vascular dementia. Neurol. Res. 2018, 40, 68–77. [Google Scholar] [CrossRef]
  240. Roman, G.C. Cholinergic dysfunction in vascular dementia. Curr. Psychiatry Rep. 2005, 7, 18–26. [Google Scholar] [CrossRef]
  241. Wang, J.; Zhang, H.Y.; Tang, X.C. Cholinergic deficiency involved in vascular dementia: Possible mechanism and strategy of treatment. Acta Pharmacol. Sin. 2009, 30, 879–888. [Google Scholar] [CrossRef]
  242. Mitsushima, D.; Sano, A.; Takahashi, T. A cholinergic trigger drives learning-induced plasticity at hippocampal synapses. Nat. Commun. 2013, 4, 2760. [Google Scholar] [CrossRef] [Green Version]
  243. Ribeiro, F.F.; Xapelli, S.; Miranda-Lourenco, C.; Tanqueiro, S.R.; Fonseca-Gomes, J.; Diogenes, M.J.; Ribeiro, J.A.; Sebastiao, A.M. Purine nucleosides in neuroregeneration and neuroprotection. Neuropharmacology 2016, 104, 226–242. [Google Scholar] [CrossRef] [PubMed]
  244. Mouro, F.M.; Rombo, D.M.; Dias, R.B.; Ribeiro, J.A.; Sebastiao, A.M. Adenosine A2A receptors facilitate synaptic NMDA currents in CA1 pyramidal neurons. Br. J. Pharmacol. 2018, 175, 4386–4397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Sreedharan, J.; Blair, I.P.; Tripathi, V.B.; Hu, X.; Vance, C.; Rogelj, B.; Ackerley, S.; Durnall, J.C.; Williams, K.L.; Buratti, E.; et al. TDP-43 Mutations in Familial and Sporadic Amyotrophic Lateral Sclerosis. Science 2008, 319, 1668–1672. [Google Scholar] [CrossRef]
  246. Alami, N.; Smith, R.B.; Carrasco, M.A.; Williams, L.A.; Winborn, C.S.; Han, S.S.; Kiskinis, E.; Winborn, B.; Freibaum, B.D.; Kanagaraj, A.; et al. Axonal Transport of TDP-43 mRNA Granules Is Impaired by ALS-Causing Mutations. Neuron 2014, 81, 536–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Polymenidou, M.; Lagier-Tourenne, C.; Hutt, K.R.; Huelga, S.C.; Moran, J.; Liang, T.Y.; Ling, S.-C.; Sun, E.; Wancewicz, E.; Mazur, C.; et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat. Neurosci. 2011, 14, 459–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Costessi, L.; Porro, F.; Iaconcig, A.; Muro, A.F. TDP-43 regulates beta-adducin (Add2) transcript stability. RNA Biol. 2014, 11, 1280–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Cembrowski, M.S.; Spruston, N. Heterogeneity within classical cell types is the rule: Lessons from hippocampal pyramidal neurons. Nat. Rev. Neurosci. 2019, 20, 193–204. [Google Scholar] [CrossRef]
  250. Dalton, M.A.; Zeidman, P.; Barry, D.N.; Williams, E.; Maguire, E.A. Segmenting subregions of the human hippocampus on structural magnetic resonance image scans: An illustrated tutorial. Brain Neurosci. Adv. 2017, 1, 2398212817701448. [Google Scholar] [CrossRef]
  251. Spruston, N. Pyramidal neurons: Dendritic structure and synaptic integration. Nat. Rev. Neurosci. 2008, 9, 206–221. [Google Scholar] [CrossRef]
  252. Claiborne, B.J.; Amaral, D.G.; Cowan, W.M. Quantitative, three-dimensional analysis of granule cell dendrites in the rat dentate gyrus. J. Comp. Neurol. 1990, 302, 206–219. [Google Scholar] [CrossRef]
  253. Kumar, V.; Zhang, M.X.; Swank, M.W.; Kunz, J.; Wu, G.Y. Regulation of dendritic morphogenesis by Ras-PI3K-Akt-mTOR and Ras-MAPK signaling pathways. J. Neurosci. 2005, 25, 11288–11299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Maletic-Savatic, M.; Malinow, R.; Svoboda, K. Rapid dendritic morphogenesis in CA1 hippocampal dendrites induced by synaptic activity. Science 1999, 283, 1923–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Cline, H.T. Dendritic arbor development and synaptogenesis. Curr. Opin. Neurobiol. 2001, 11, 118–126. [Google Scholar] [CrossRef]
  256. Dailey, M.E.; Smith, S.J. The dynamics of dendritic structure in developing hippocampal slices. J. Neurosci. 1996, 16, 2983–2994. [Google Scholar] [CrossRef] [PubMed]
  257. Parrish, J.Z.; Emoto, K.; Kim, M.D.; Jan, Y.N. Mechanisms that regulate establishment, maintenance, and remodeling of dendritic fields. Annu. Rev. Neurosci. 2007, 30, 399–423. [Google Scholar] [CrossRef]
  258. Sweet, E.S.; Tseng, C.-Y.; Firestein, B. To branch or not to branch: How PSD-95 regulates dendrites and spines. Bioarchitecture 2011, 1, 69–73. [Google Scholar] [CrossRef] [Green Version]
  259. Tata, D.A.; Anderson, B.J. The effects of chronic glucocorticoid exposure on dendritic length, synapse numbers and glial volume in animal models: Implications for hippocampal volume reductions in depression. Physiol. Behav. 2010, 99, 186–193. [Google Scholar] [CrossRef]
  260. Conrad, C.D.; McLaughlin, K.J.; Harman, J.S.; Foltz, C.; Wieczorek, L.; Lightner, E.; Wright, R.L. Chronic glucocorticoids increase hippocampal vulnerability to neurotoxicity under conditions that produce CA3 dendritic retraction but fail to impair spatial recognition memory. J. Neurosci. 2007, 27, 8278–8285. [Google Scholar] [CrossRef] [Green Version]
  261. Tolwani, R.J.; Buckmaster, P.S.; Varma, S.; Cosgaya, J.M.; Wu, Y.; Suri, C.; Shooter, E.M. BDNF overexpression increases dendrite complexity in hippocampal dentate gyrus. Neuroscience 2002, 114, 795–805. [Google Scholar] [CrossRef]
  262. Kulkarni, V.A.; Firestein, B.L. The dendritic tree and brain disorders. Mol. Cell Neurosci. 2012, 50, 10–20. [Google Scholar] [CrossRef]
  263. Kolomeets, N.S.; Orlovskaya, D.D.; Uranova, N.A. Decreased numerical density of CA3 hippocampal mossy fiber synapses in schizophrenia. Synapse 2007, 61, 615–621. [Google Scholar] [CrossRef] [PubMed]
  264. Anderton, B.H.; Callahan, L.; Coleman, P.; Davies, P.; Flood, D.; Jicha, G.A.; Ohm, T.; Weaver, C. Dendritic changes in Alzheimer’s disease and factors that may underlie these changes. Prog. Neurobiol. 1998, 55, 595–609. [Google Scholar] [CrossRef]
  265. Sousa, N.; Lukoyanov, N.; Madeira, M.; Almeida, O.; Paula-Barbosa, M. Reorganization of the morphology of hippocampal neurites and synapses after stress-induced damage correlates with behavioral improvement. Neuroscience 2000, 97, 253–266. [Google Scholar] [CrossRef]
  266. Sorra, K.E.; Harris, K.M. Overview on the structure, composition, function, development, and plasticity of hippocampal dendritic spines. Hippocampus 2000, 10, 501–511. [Google Scholar] [CrossRef]
  267. Ultanir, S.K.; Kim, J.E.; Hall, B.J.; Deerinck, T.; Ellisman, M.; Ghosh, A. Regulation of spine morphology and spine density by NMDA receptor signaling in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 19553–19558. [Google Scholar] [CrossRef] [Green Version]
  268. von Bohlen Und Halbach, O. Structure and function of dendritic spines within the hippocampus. Ann. Anat. 2009, 191, 518–531. [Google Scholar] [CrossRef]
  269. Kasai, H.; Matsuzaki, M.; Noguchi, J.; Yasumatsu, N.; Nakahara, H. Structure-stability-function relationships of dendritic spines. Trends Neurosci. 2003, 26, 360–368. [Google Scholar] [CrossRef]
  270. Bourne, J.N.; Harris, K.M. Balancing structure and function at hippocampal dendritic spines. Annu. Rev. Neurosci. 2008, 31, 47–67. [Google Scholar] [CrossRef] [Green Version]
  271. Bourne, J.; Harris, K.M. Do thin spines learn to be mushroom spines that remember? Curr. Opin. Neurobiol. 2007, 17, 381–386. [Google Scholar] [CrossRef]
  272. Trachtenberg, J.T.; Chen, B.E.; Knott, G.W.; Feng, G.; Sanes, J.R.; Welker, E.; Svoboda, K. Long-term in vivo imaging of experience-dependent synaptic plasticity in adult cortex. Nature 2002, 420, 788–794. [Google Scholar] [CrossRef]
  273. Skoff, R.P.; Hamburger, V. Fine structure of dendritic and axonal growth cones in embryonic chick spinal cord. J. Comp. Neurol. 1974, 153, 107–147. [Google Scholar] [CrossRef] [PubMed]
  274. Lohmann, C.; Bonhoeffer, T. A role for local calcium signaling in rapid synaptic partner selection by dendritic filopodia. Neuron 2008, 59, 253–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Sorra, K.E.; Fiala, J.C.; Harris, K.M. Critical assessment of the involvement of perforations, spinules, and spine branching in hippocampal synapse formation. J. Comp. Neurol. 1998, 398, 225–240. [Google Scholar] [CrossRef]
  276. Leung, C.C.Y.; Wong, Y.H. Role of G Protein-Coupled Receptors in the Regulation of Structural Plasticity and Cognitive Function. Molecules 2017, 22, 1239. [Google Scholar] [CrossRef] [Green Version]
  277. Tsai, J.; Grutzendler, J.; Duff, K.; Gan, W.B. Fibrillar amyloid deposition leads to local synaptic abnormalities and breakage of neuronal branches. Nat. Neurosci. 2004, 7, 1181–1183. [Google Scholar] [CrossRef]
  278. Bellot, A.; Guivernau, B.; Tajes, M.; Bosch-Morato, M.; Valls-Comamala, V.; Munoz, F.J. The structure and function of actin cytoskeleton in mature glutamatergic dendritic spines. Brain Res. 2014, 1573, 1–16. [Google Scholar] [CrossRef] [Green Version]
  279. Einstein, G.; Buranosky, R.; Crain, B.J. Dendritic pathology of granule cells in Alzheimer’s disease is unrelated to neuritic plaques. J. Neurosci. 1994, 14, 5077–5088. [Google Scholar] [CrossRef]
  280. Moolman, D.L.; Vitolo, O.V.; Vonsattel, J.P.; Shelanski, M.L. Dendrite and dendritic spine alterations in Alzheimer models. J. Neurocytol. 2004, 33, 377–387. [Google Scholar] [CrossRef]
  281. Sun, G.Z.; He, Y.C.; Ma, X.K.; Li, S.T.; Chen, D.J.; Gao, M.; Qiu, S.F.; Yin, J.X.; Shi, J.; Wu, J. Hippocampal synaptic and neural network deficits in young mice carrying the human APOE4 gene. CNS Neurosci. Ther. 2017, 23, 748–758. [Google Scholar] [CrossRef] [Green Version]
  282. Kao, Y.C.; Wang, I.F.; Tsai, K.J. miRNA-34c Overexpression Causes Dendritic Loss and Memory Decline. Int. J. Mol. Sci. 2018, 19, 2323. [Google Scholar] [CrossRef] [Green Version]
  283. Pristerà, A.; Saraulli, D.; Vecchioli, S.F.; Strimpakos, G.; Costanzi, M.; di Certo, M.G.; Cannas, S.; Ciotti, M.T.; Tirone, F.; Mattei, E.; et al. Impact of N-tau on adult hippocampal neurogenesis, anxiety, and memory. Neurobiol. Aging 2013, 34, 2551–2563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Androuin, A.; Potier, B.; Nägerl, U.V.; Cattaert, D.; Danglot, L.; Thierry, M.; Youssef, I.; Triller, A.; Duyckaerts, C.; El Hachimi, K.H.; et al. Evidence for altered dendritic spine compartmentalization in Alzheimer’s disease and functional effects in a mouse model. Acta Neuropathol. 2018, 135, 839–854. [Google Scholar] [CrossRef] [PubMed]
  285. Zhang, H.; Wu, L.; Pchitskaya, E.; Zakharova, O.; Saito, T.; Saido, T.; Bezprozvanny, I. Neuronal Store-Operated Calcium Entry and Mushroom Spine Loss in Amyloid Precursor Protein Knock-In Mouse Model of Alzheimer’s Disease. J. Neurosci. 2015, 35, 13275–13286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Dachsel, J.C.; Behrouz, B.; Yue, M.; Beevers, J.E.; Melrose, H.L.; Farrer, M.J. A comparative study of Lrrk2 function in primary neuronal cultures. Park. Relat. Disord. 2010, 16, 650–655. [Google Scholar] [CrossRef] [Green Version]
  287. Sepulveda, B.; Mesias, R.; Li, X.; Yue, Z.; Benson, D.L. Short- and long-term effects of LRRK2 on axon and dendrite growth. PLoS ONE 2013, 8, e61986. [Google Scholar] [CrossRef] [Green Version]
  288. Winner, B.; Melrose, H.; Zhao, C.; Hinkle, K.; Yue, M.; Kent, C.; Braithwaite, A.; Ogholikhan, S.; Aigner, R.; Winkler, J. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol. Dis. 2011, 41, 706–716. [Google Scholar] [CrossRef] [Green Version]
  289. Weerasinghe-Mudiyanselage, P.D.E.; Ang, M.J.; Wada, M.; Kim, S.H.; Shin, T.; Yang, M.; Moon, C. Acute MPTP Treatment Impairs Dendritic Spine Density in the Mouse Hippocampus. Brain Sci. 2021, 11, 833. [Google Scholar] [CrossRef]
  290. Froula, J.M.; Henderson, B.W.; Gonzalez, J.C.; Vaden, J.H.; McLean, J.W.; Wu, Y.; Banumurthy, G.; Overstreet-Wadiche, L.; Herskowitz, J.H.; Volpicelli-Daley, L.A. alpha-Synuclein fibril-induced paradoxical structural and functional defects in hippocampal neurons. Acta Neuropathol. Commun. 2018, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  291. da Silva, M.O.; Santejo, M.; Babcock, I.; Magalhães, A.; Minamide, L.; Castillo, E.; Gerhardt, E.; Fahlbusch, C.; Swanson, R.; Outeiro, T. Cofilin pathology is a new player on α-synuclein-induced spine impairment in models of hippocampal synucleinopathy. Biorxiv 2021. [Google Scholar] [CrossRef]
  292. Anglada-Huguet, M.; Vidal-Sancho, L.; Giralt, A.; Barriga, G.G.-D.; Xifró, X.; Alberch, J. Prostaglandin E2 EP2 activation reduces memory decline in R6/1 mouse model of Huntington’s disease by the induction of BDNF-dependent synaptic plasticity. Neurobiol. Dis. 2016, 95, 22–34. [Google Scholar] [CrossRef]
  293. Milnerwood, A.J.; Parsons, M.P.; Young, F.B.; Singaraja, R.R.; Franciosi, S.; Volta, M.; Bergeron, S.; Hayden, M.R.; Raymond, L.A. Memory and synaptic deficits in Hip14/DHHC17 knockout mice. Proc. Natl. Acad. Sci. USA 2013, 110, 20296–20301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Miguez, A.; Garcia-Diaz Barriga, G.; Brito, V.; Straccia, M.; Giralt, A.; Gines, S.; Canals, J.M.; Alberch, J. Fingolimod (FTY720) enhances hippocampal synaptic plasticity and memory in Huntington’s disease by preventing p75NTR up-regulation and astrocyte-mediated inflammation. Hum. Mol. Genet. 2015, 24, 4958–4970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Nithianantharajah, J.; Barkus, C.; Murphy, M.; Hannan, A.J. Gene-environment interactions modulating cognitive function and molecular correlates of synaptic plasticity in Huntington’s disease transgenic mice. Neurobiol. Dis. 2008, 29, 490–504. [Google Scholar] [CrossRef]
  296. Crombe, A.; Planche, V.; Raffard, G.; Bourel, J.; Dubourdieu, N.; Panatier, A.; Fukutomi, H.; Dousset, V.; Oliet, S.; Hiba, B.; et al. Deciphering the microstructure of hippocampal subfields with in vivo DTI and NODDI: Applications to experimental multiple sclerosis. NeuroImage 2018, 172, 357–368. [Google Scholar] [CrossRef] [PubMed]
  297. Planche, V.; Panatier, A.; Hiba, B.; Ducourneau, E.-G.; Raffard, G.; Dubourdieu, N.; Maitre, M.; Lesté-Lasserre, T.; Brochet, B.; Dousset, V.; et al. Selective dentate gyrus disruption causes memory impairment at the early stage of experimental multiple sclerosis. Brain Behav. Immun. 2017, 60, 240–254. [Google Scholar] [CrossRef] [PubMed]
  298. Baltan, S.; Jawaid, S.S.; Chomyk, A.M.; Kidd, G.J.; Chen, J.; Battapady, H.D.; Chan, R.; Dutta, R.; Trapp, B.D. Neuronal hibernation following hippocampal demyelination. Acta Neuropathol. Commun. 2021, 9, 34. [Google Scholar] [CrossRef]
  299. Chen, X.; Jiang, X.-M.; Zhao, L.-J.; Sun, L.-L.; Yan, M.-L.; Tian, Y.; Zhang, S.; Duan, M.-J.; Zhao, H.-M.; Li, W.-R.; et al. MicroRNA-195 prevents dendritic degeneration and neuron death in rats following chronic brain hypoperfusion. Cell Death Dis. 2017, 8, e2850. [Google Scholar] [CrossRef] [Green Version]
  300. Guo, S.; Xu, J.-J.; Wei, N.; Han, J.-Y.; Xue, R.; Xu, P.-S.; Gao, C.-Y. Honokiol Attenuates the Memory Impairments, Oxidative Stress, Neuroinflammation, and GSK-3 β Activation in Vascular Dementia Rats. J. Alzheimer’s Dis. 2019, 71, 97–108. [Google Scholar] [CrossRef]
  301. Zhu, Y.; Zhang, Q.; Zhang, W.; Li, N.; Dai, Y.; Tu, J.; Yang, F.; Brann, D.W.; Wang, R. Protective effect of 17β-estradiol upon hippocampal spine density and cognitive function in an animal model of vascular dementia. Sci. Rep. 2017, 7, 42660. [Google Scholar] [CrossRef] [Green Version]
  302. Jian, W.X.; Zhang, Z.; Zhan, J.H.; Chu, S.F.; Peng, Y.; Zhao, M.; Wang, Q.; Chen, N.H. Donepezil attenuates vascular dementia in rats through increasing BDNF induced by reducing HDAC6 nuclear translocation. Acta Pharmacol. Sin. 2020, 41, 588–598. [Google Scholar] [CrossRef]
  303. Fogarty, M.J.; Mu, E.W.H.; Lavidis, N.A.; Noakes, P.G.; Bellingham, M.C. Motor Areas Show Altered Dendritic Structure in an Amyotrophic Lateral Sclerosis Mouse Model. Front Neurosci. 2017, 11, 609. [Google Scholar] [CrossRef] [PubMed]
  304. Jack, C.R., Jr.; Knopman, D.S.; Jagust, W.J.; Shaw, L.M.; Aisen, P.S.; Weiner, M.W.; Petersen, R.C.; Trojanowski, J.Q. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9, 119–128. [Google Scholar] [CrossRef] [Green Version]
  305. Blennow, K.; Hampel, H.; Weiner, M.; Zetterberg, H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat. Rev. Neurol. 2010, 6, 131–144. [Google Scholar] [CrossRef] [PubMed]
  306. Nisbet, R.M.; Polanco, J.C.; Ittner, L.M.; Gotz, J. Tau aggregation and its interplay with amyloid-beta. Acta Neuropathol. 2015, 129, 207–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Canevari, L.; Abramov, A.Y.; Duchen, M.R. Toxicity of amyloid beta peptide: Tales of calcium, mitochondria, and oxidative stress. Neurochem. Res. 2004, 29, 637–650. [Google Scholar] [CrossRef] [PubMed]
  308. Mattson, M.P.; Chan, S.L. Neuronal and glial calcium signaling in Alzheimer’s disease. Cell Calcium. 2003, 34, 385–397. [Google Scholar] [CrossRef]
  309. Lee, G.; Neve, R.L.; Kosik, K.S. The microtubule binding domain of tau protein. Neuron 1989, 2, 1615–1624. [Google Scholar] [CrossRef]
  310. Kent, S.A.; Spires-Jones, T.L.; Durrant, C.S. The physiological roles of tau and Abeta: Implications for Alzheimer’s disease pathology and therapeutics. Acta Neuropathol. 2020, 140, 417–447. [Google Scholar] [CrossRef]
  311. Spires-Jones, T.; Knafo, S. Spines, plasticity, and cognition in Alzheimer’s model mice. Neural. Plast. 2012, 2012, 319836. [Google Scholar] [CrossRef]
  312. Toni, N.; Sultan, S. Synapse formation on adult-born hippocampal neurons. Eur. J. Neurosci. 2011, 33, 1062–1068. [Google Scholar] [CrossRef]
  313. Gong, X.; Lu, X.; Zhan, L.; Sui, H.; Qi, X.; Ji, Z.; Niu, X.; Liu, L. Role of the SNK-SPAR pathway in the development of Alzheimer’s disease. IUBMB Life 2010, 62, 214–221. [Google Scholar] [CrossRef] [PubMed]
  314. Hoover, B.R.; Reed, M.N.; Su, J.; Penrod, R.D.; Kotilinek, L.A.; Grant, M.K.; Pitstick, R.; Carlson, G.A.; Lanier, L.M.; Yuan, L.-L.; et al. Tau Mislocalization to Dendritic Spines Mediates Synaptic Dysfunction Independently of Neurodegeneration. Neuron 2010, 68, 1067–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Pluta, R.; Ułamek-Kozioł, M. Tau Protein-Targeted Therapies in Alzheimer’s Disease: Current State and Future Perspectives; Exon Publications: Brisbane, Australia, 2020; pp. 69–82. [Google Scholar]
  316. Xin, S.H.; Tan, L.; Cao, X.; Yu, J.T.; Tan, L. Clearance of Amyloid Beta and Tau in Alzheimer’s Disease: From Mechanisms to Therapy. Neurotox Res. 2018, 34, 733–748. [Google Scholar] [CrossRef] [PubMed]
  317. Polanco, J.C.; Li, C.; Bodea, L.-G.; Martinez-Marmol, R.; Meunier, F.A.; Götz, J. Amyloid-β and tau complexity—towards improved biomarkers and targeted therapies. Nat. Rev. Neurol. 2018, 14, 22–39. [Google Scholar] [CrossRef]
  318. McNeill, T.H.; Brown, S.A.; Rafols, J.A.; Shoulson, I. Atrophy of medium spiny I striatal dendrites in advanced Parkinson’s disease. Brain Res. 1988, 455, 148–152. [Google Scholar] [CrossRef]
  319. Day, M.; Wang, Z.; Ding, J.; An, X.; Ingham, C.A.; Shering, A.F.; Wokosin, D.; Ilijic, E.; Sun, Z.; Sampson, A.R.; et al. Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nat. Neurosci. 2006, 9, 251–259. [Google Scholar] [CrossRef]
  320. Zaja-Milatovic, S.; Milatovic, D.; Schantz, A.M.; Zhang, J.; Montine, K.S.; Samii, A.; Deutch, A.Y.; Montine, T.J. Dendritic degeneration in neostriatal medium spiny neurons in Parkinson disease. Neurology 2005, 64, 545–547. [Google Scholar] [CrossRef]
  321. Stephens, B.; Mueller, A.; Shering, A.; Hood, S.; Taggart, P.; Arbuthnott, G.; Bell, J.; Kilford, L.; Kingsbury, A.; Daniel, S.; et al. Evidence of a breakdown of corticostriatal connections in Parkinson’s disease. Neuroscience 2005, 132, 741–754. [Google Scholar] [CrossRef]
  322. Ingham, C.A.; Hood, S.H.; Arbuthnott, G.W. Spine density on neostriatal neurones changes with 6-hydroxydopamine lesions and with age. Brain Res. 1989, 503, 334–338. [Google Scholar] [CrossRef]
  323. Borgkvist, A.; Malmlof, T.; Feltmann, K.; Lindskog, M.; Schilstrom, B. Dopamine in the hippocampus is cleared by the norepinephrine transporter. Int. J. Neuropsychopharmacol. 2012, 15, 531–540. [Google Scholar] [CrossRef] [Green Version]
  324. Park, J.H.; Enikolopov, G. Transient elevation of adult hippocampal neurogenesis after dopamine depletion. Exp. Neurol. 2010, 222, 267–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Gangarossa, G.; Longueville, S.; De Bundel, D.; Perroy, J.; Herve, D.; Girault, J.A.; Valjent, E. Characterization of dopamine D1 and D2 receptor-expressing neurons in the mouse hippocampus. Hippocampus 2012, 22, 2199–2207. [Google Scholar] [CrossRef] [PubMed]
  326. Shinohara, R.; Taniguchi, M.; Ehrlich, A.T.; Yokogawa, K.; Deguchi, Y.; Cherasse, Y.; Lazarus, M.; Urade, Y.; Ogawa, A.; Kitaoka, S.; et al. Dopamine D1 receptor subtype mediates acute stress-induced dendritic growth in excitatory neurons of the medial prefrontal cortex and contributes to suppression of stress susceptibility in mice. Mol. Psychiatry 2018, 23, 1717–1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Neuner, J.; Ovsepian, S.V.; Dorostkar, M.; Filser, S.; Gupta, A.; Michalakis, S.; Biel, M.; Herms, J. Pathological alpha-synuclein impairs adult-born granule cell development and functional integration in the olfactory bulb. Nat. Commun. 2014, 5, 3915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  328. Koch, J.C.; Bitow, F.; Haack, J.; D’Hedouville, Z.; Zhang, J.-N.; Tönges, L.; Michel, U.; Oliveira, L.M.A.; Jovin, T.M.; Liman, J.; et al. Alpha-Synuclein affects neurite morphology, autophagy, vesicle transport and axonal degeneration in CNS neurons. Cell Death Dis. 2015, 6, e1811. [Google Scholar] [CrossRef] [Green Version]
  329. Galvin, J.E.; Uryu, K.; Lee, V.M.; Trojanowski, J.Q. Axon pathology in Parkinson’s disease and Lewy body dementia hippocampus contains alpha-, beta-, and gamma-synuclein. Proc. Natl. Acad. Sci. USA 1999, 96, 13450–13455. [Google Scholar] [CrossRef] [Green Version]
  330. Scatton, B.; Javoy-Agid, F.; Rouquier, L.; Dubois, B.; Agid, Y. Reduction of cortical dopamine, noradrenaline, serotonin and their metabolites in Parkinson’s disease. Brain Res. 1983, 275, 321–328. [Google Scholar] [CrossRef]
  331. Deusser, J.; Schmidt, S.; Ettle, B.; Plotz, S.; Huber, S.; Muller, C.P.; Masliah, E.; Winkler, J.; Kohl, Z. Serotonergic dysfunction in the A53T alpha-synuclein mouse model of Parkinson’s disease. J. Neurochem. 2015, 135, 589–597. [Google Scholar] [CrossRef] [Green Version]
  332. Trakhtenberg, E.F.; Goldberg, J.L. The role of serotonin in axon and dendrite growth. Int. Rev. Neurobiol. 2012, 106, 105–126. [Google Scholar] [CrossRef]
  333. Lindgren, H.S.; Dunnett, S.B. Cognitive dysfunction and depression in Parkinson’s disease: What can be learned from rodent models? Eur. J. Neurosci. 2012, 35, 1894–1907. [Google Scholar] [CrossRef]
  334. Ji, Y.; Pang, P.T.; Feng, L.; Lu, B. Cyclic AMP controls BDNF-induced TrkB phosphorylation and dendritic spine formation in mature hippocampal neurons. Nat. Neurosci. 2005, 8, 164–172. [Google Scholar] [CrossRef] [PubMed]
  335. Norrholm, S.D.; Ouimet, C.C. Chronic fluoxetine administration to juvenile rats prevents age-associated dendritic spine proliferation in hippocampus. Brain Res. 2000, 883, 205–215. [Google Scholar] [CrossRef]
  336. Bender, H.; Fietz, S.A.; Richter, F.; Stanojlovic, M. Alpha-Synuclein Pathology Coincides With Increased Number of Early Stage Neural Progenitors in the Adult Hippocampus. Front. Cell Dev. Biol. 2021, 9, 691560. [Google Scholar] [CrossRef]
  337. Alim, M.A.; Ma, Q.-L.; Takeda, K.; Aizawa, T.; Matsubara, M.; Nakamura, M.; Asada, A.; Saito, T.; Xkaji, M.; Yoshii, M.; et al. Demonstration of a role for α-synuclein as a functional microtubule-associated protein. J. Alzheimer’s Dis. 2004, 6, 435–442. [Google Scholar] [CrossRef] [PubMed]
  338. Bonini, N.M.; Giasson, B.I. Snaring the function of alpha-synuclein. Cell 2005, 123, 359–361. [Google Scholar] [CrossRef] [PubMed]
  339. Stefanis, L. alpha-Synuclein in Parkinson’s disease. Cold Spring Harb. Perspect Med. 2012, 2, a009399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  340. Wersinger, C.; Rusnak, M.; Sidhu, A. Modulation of the trafficking of the human serotonin transporter by human alpha-synuclein. Eur. J. Neurosci. 2006, 24, 55–64. [Google Scholar] [CrossRef] [PubMed]
  341. Chaji, D.; Venkatesh, V.S.; Shirao, T.; Day, D.J.; Ellenbroek, B.A. Genetic Knockout of the Serotonin Reuptake Transporter Results in the Reduction of Dendritic Spines in In vitro Rat Cortical Neuronal Culture. J. Mol. Neurosci. 2021, 71, 2210–2218. [Google Scholar] [CrossRef]
  342. Nagano-Saito, A.; Habak, C.; Mejía-Constaín, B.; Degroot, C.; Monetta, L.; Jubault, T.; Bedetti, C.; Lafontaine, A.-L.; Chouinard, S.; Soland, V.; et al. Effect of mild cognitive impairment on the patterns of neural activity in early Parkinson’s disease. Neurobiol. Aging 2014, 35, 223–231. [Google Scholar] [CrossRef]
  343. Milnerwood, A.J.; Raymond, L.A. Early synaptic pathophysiology in neurodegeneration: Insights from Huntington’s disease. Trends Neurosci. 2010, 33, 513–523. [Google Scholar] [CrossRef]
  344. Raymond, L.A.; Andre, V.M.; Cepeda, C.; Gladding, C.M.; Milnerwood, A.J.; Levine, M.S. Pathophysiology of Huntington’s disease: Time-dependent alterations in synaptic and receptor function. Neuroscience 2011, 198, 252–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Schmidt, M.E.; Buren, C.; Mackay, J.P.; Cheung, D.; Dal Cengio, L.; Raymond, L.A.; Hayden, M.R. Altering cortical input unmasks synaptic phenotypes in the YAC128 cortico-striatal co-culture model of Huntington disease. BMC Biol. 2018, 16, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  346. Carmo, C.; Naia, L.; Lopes, C.; Rego, A.C. Mitochondrial Dysfunction in Huntington’s Disease. Adv. Exp. Med. Biol. 2018, 1049, 59–83. [Google Scholar] [CrossRef] [PubMed]
  347. Saavedra, A.; Garcia-Diaz Barriga, G.; Perez-Navarro, E.; Alberch, J. Huntington’s disease: Novel therapeutic perspectives hanging in the balance. Expert Opin. Ther. Targets 2018, 22, 385–399. [Google Scholar] [CrossRef]
  348. Li, X.J.; Orr, A.L.; Li, S. Impaired mitochondrial trafficking in Huntington’s disease. Biochim. Biophys. Acta 2010, 1802, 62–65. [Google Scholar] [CrossRef] [Green Version]
  349. Rangaraju, V.; Lewis, T.L., Jr.; Hirabayashi, Y.; Bergami, M.; Motori, E.; Cartoni, R.; Kwon, S.K.; Courchet, J. Pleiotropic Mitochondria: The Influence of Mitochondria on Neuronal Development and Disease. J. Neurosci. 2019, 39, 8200–8208. [Google Scholar] [CrossRef] [Green Version]
  350. Steib, K.; Schaffner, I.; Jagasia, R.; Ebert, B.; Lie, D.C. Mitochondria modify exercise-induced development of stem cell-derived neurons in the adult brain. J. Neurosci. 2014, 34, 6624–6633. [Google Scholar] [CrossRef]
  351. Magistretti, P.J.; Allaman, I. A cellular perspective on brain energy metabolism and functional imaging. Neuron 2015, 86, 883–901. [Google Scholar] [CrossRef] [Green Version]
  352. Gauthier, L.R.; Charrin, B.C.; Borrell, M.; Dompierre, J.P.; Rangone, H.; Cordelières, F.; De Mey, J.; MacDonald, M.E.; Leßmann, V.; Humbert, S.; et al. Huntingtin Controls Neurotrophic Support and Survival of Neurons by Enhancing BDNF Vesicular Transport along Microtubules. Cell 2004, 118, 127–138. [Google Scholar] [CrossRef] [Green Version]
  353. McAllister, A.K. Cellular and molecular mechanisms of dendrite growth. Cereb. Cortex. 2000, 10, 963–973. [Google Scholar] [CrossRef] [Green Version]
  354. Labbadia, J.; Morimoto, R.I. Huntington’s disease: Underlying molecular mechanisms and emerging concepts. Trends Biochem. Sci. 2013, 38, 378–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  355. Zuccato, C.; Valenza, M.; Cattaneo, E. Molecular mechanisms and potential therapeutical targets in Huntington’s disease. Physiol. Rev. 2010, 90, 905–981. [Google Scholar] [CrossRef]
  356. Flippo, K.H.; Strack, S. Mitochondrial dynamics in neuronal injury, development and plasticity. J. Cell Sci. 2017, 130, 671–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  357. Li, Z.; Okamoto, K.; Hayashi, Y.; Sheng, M. The importance of dendritic mitochondria in the morphogenesis and plasticity of spines and synapses. Cell 2004, 119, 873–887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  358. Shirendeb, U.P.; Calkins, M.J.; Manczak, M.; Anekonda, V.; Dufour, B.; McBride, J.L.; Mao, P.; Reddy, P.H. Mutant huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum. Mol. Genet. 2012, 21, 406–420. [Google Scholar] [CrossRef] [PubMed]
  359. Ettcheto, M.; Busquets, O.; Cano, A.; Sánchez-Lopez, E.; Manzine, P.R.; Espinosa-Jimenez, T.; Verdaguer, E.; Sureda, F.X.; Olloquequi, J.; Castro-Torres, R.D.; et al. Pharmacological Strategies to Improve Dendritic Spines in Alzheimer’s Disease. J. Alzheimer’s Dis. 2021, 82, S91–S107. [Google Scholar] [CrossRef]
  360. Grussu, F.; Schneider, T.; Tur, C.; Yates, R.L.; Tachrount, M.; Ianus, A.; Yiannakas, M.C.; Newcombe, J.; Zhang, H.; Alexander, D.C.; et al. Neurite dispersion: A new marker of multiple sclerosis spinal cord pathology? Ann. Clin. Transl. Neurol. 2017, 4, 663–679. [Google Scholar] [CrossRef]
  361. Gilmore, C.P.; DeLuca, G.C.; Bo, L.; Owens, T.; Lowe, J.; Esiri, M.M.; Evangelou, N. Spinal cord atrophy in multiple sclerosis caused by white matter volume loss. Arch Neurol. 2005, 62, 1859–1862. [Google Scholar] [CrossRef] [Green Version]
  362. D’Souza, S.; Alinauskas, K.; McCrea, E.; Goodyer, C.; Antel, J.P. Differential susceptibility of human CNS-derived cell populations to TNF-dependent and independent immune-mediated injury. J. Neurosci. 1995, 15, 7293–7300. [Google Scholar] [CrossRef]
  363. Akassoglou, K.; Bauer, J.; Kassiotis, G.; Pasparakis, M.; Lassmann, H.; Kollias, G.; Probert, L. Oligodendrocyte apoptosis and primary demyelination induced by local TNF/p55TNF receptor signaling in the central nervous system of transgenic mice: Models for multiple sclerosis with primary oligodendrogliopathy. Am. J. Pathol. 1998, 153, 801–813. [Google Scholar] [CrossRef]
  364. Andrews, T.; Zhang, P.; Bhat, N.R. TNFalpha potentiates IFNgamma-induced cell death in oligodendrocyte progenitors. J. Neurosci. Res. 1998, 54, 574–583. [Google Scholar] [CrossRef]
  365. Ben-Hur, T.; Ben-Menachem, O.; Furer, V.; Einstein, O.; Mizrachi-Kol, R.; Grigoriadis, N. Effects of proinflammatory cytokines on the growth, fate, and motility of multipotential neural precursor cells. Mol. Cell Neurosci. 2003, 24, 623–631. [Google Scholar] [CrossRef]
  366. Yoo, S.W.; Motari, M.G.; Susuki, K.; Prendergast, J.; Mountney, A.; Hurtado, A.; Schnaar, R.L. Sialylation regulates brain structure and function. FASEB J. 2015, 29, 3040–3053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Wang, Q.; Yu, S.; Simonyi, A.; Sun, G.Y.; Sun, A.Y. Kainic acid-mediated excitotoxicity as a model for neurodegeneration. Molecular. Neurobiol. 2005, 31, 3–16. [Google Scholar] [CrossRef]
  368. Vallejo-Illarramendi, A.; Domercq, M.; Perez-Cerda, F.; Ravid, R.; Matute, C. Increased expression and function of glutamate transporters in multiple sclerosis. Neurobiol. Dis. 2006, 21, 154–164. [Google Scholar] [CrossRef]
  369. Crochemore, C.; Lu, J.; Wu, Y.; Liposits, Z.; Sousa, N.; Holsboer, F.; Almeida, O.F. Direct targeting of hippocampal neurons for apoptosis by glucocorticoids is reversible by mineralocorticoid receptor activation. Mol. Psychiatry 2005, 10, 790–798. [Google Scholar] [CrossRef] [Green Version]
  370. Lu, J.; Goula, D.; Sousa, N.; Almeida, O.F. Ionotropic and metabotropic glutamate receptor mediation of glucocorticoid-induced apoptosis in hippocampal cells and the neuroprotective role of synaptic N-methyl-D-aspartate receptors. Neuroscience 2003, 121, 123–131. [Google Scholar] [CrossRef] [Green Version]
  371. Uttner, I.; Muller, S.; Zinser, C.; Maier, M.; Süssmuth, S.; Claus, A.; Ostermann, B.; Elitok, E.; Ecker, D.; Brettschneider, J.; et al. Reversible impaired memory induced by pulsed methylprednisolone in patients with MS. Neurology 2005, 64, 1971–1973. [Google Scholar] [CrossRef]
  372. Roozendaal, B.; de Quervain, D.J. Glucocorticoid therapy and memory function: Lessons learned from basic research. Neurology 2005, 64, 184–185. [Google Scholar] [CrossRef]
  373. Brunner, R.; Schaefer, D.; Hess, K.; Parzer, P.; Resch, F.; Schwab, S. Effect of corticosteroids on short-term and long-term memory. Neurology 2005, 64, 335–337. [Google Scholar] [CrossRef]
  374. Zhang, Y.; Zhang, H.; Wang, L.; Jiang, W.; Xu, H.; Xiao, L.; Bi, X.; Wang, J.; Zhu, S.; Zhang, R.; et al. Quetiapine enhances oligodendrocyte regeneration and myelin repair after cuprizone-induced demyelination. Schizophr. Res. 2012, 138, 8–17. [Google Scholar] [CrossRef] [PubMed]
  375. Abe, H.; Tanaka, T.; Kimura, M.; Mizukami, S.; Saito, F.; Imatanaka, N.; Akahori, Y.; Yoshida, T.; Shibutani, M. Cuprizone decreases intermediate and late-stage progenitor cells in hippocampal neurogenesis of rats in a framework of 28-day oral dose toxicity study. Toxicol. Appl. Pharmacol. 2015, 287, 210–221. [Google Scholar] [CrossRef] [PubMed]
  376. Bitzer-Quintero, O.K.; Gonzalez-Burgos, I. Immune system in the brain: A modulatory role on dendritic spine morphophysiology? Neural. Plast. 2012, 2012, 348642. [Google Scholar] [CrossRef] [PubMed]
  377. Donato, F.; Jacobsen, R.I.; Moser, M.B.; Moser, E.I. Stellate cells drive maturation of the entorhinal-hippocampal circuit. Science 2017, 355, eaai8178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  378. Song, J.; Olsen, R.H.; Sun, J.; Ming, G.L.; Song, H. Neuronal Circuitry Mechanisms Regulating Adult Mammalian Neurogenesis. Cold Spring Harb. Perspect. Biol. 2016, 8, a018937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  379. Peterson, J.W.; Bo, L.; Mork, S.; Chang, A.; Trapp, B.D. Transected neurites, apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions. Ann. Neurol. 2001, 50, 389–400. [Google Scholar] [CrossRef]
  380. Peterson, L.K.; Fujinami, R.S. Inflammation, demyelination, neurodegeneration and neuroprotection in the pathogenesis of multiple sclerosis. J. Neuroimmunol. 2007, 184, 37–44. [Google Scholar] [CrossRef] [Green Version]
  381. Spalloni, A.; Origlia, N.; Sgobio, C.; Trabalza, A.; Nutini, M.; Berretta, N.; Bernardi, G.; Domenici, L.; Ammassari-Teule, M.; Longone, P. Postsynaptic alteration of NR2A subunit and defective autophosphorylation of alphaCaMKII at threonine-286 contribute to abnormal plasticity and morphology of upper motor neurons in presymptomatic SOD1G93A mice, a murine model for amyotrophic lateral sclerosis. Cereb. Cortex. 2011, 21, 796–805. [Google Scholar]
  382. Sgobio, C.; Trabalza, A.; Spalloni, A.; Zona, C.; Carunchio, I.; Longone, P.; Ammassari-Teule, M. Abnormal medial prefrontal cortex connectivity and defective fear extinction in the presymptomatic G93A SOD1 mouse model of ALS. Genes Brain Behav. 2008, 7, 427–434. [Google Scholar] [CrossRef]
  383. Van Zundert, B.; Peuscher, M.H.; Hynynen, M.; Chen, A.; Neve, R.L.; Brown, R.H.; Constantine-Paton, M.; Bellingham, M.C. Neonatal neuronal circuitry shows hyperexcitable disturbance in a mouse model of the adult-onset neurodegenerative disease amyotrophic lateral sclerosis. J. Neurosci. 2008, 28, 10864–10874. [Google Scholar] [CrossRef]
  384. Martin, E.; Cazenave, W.; Cattaert, D.; Branchereau, P. Embryonic alteration of motoneuronal morphology induces hyperexcitability in the mouse model of amyotrophic lateral sclerosis. Neurobiol. Dis. 2013, 54, 116–126. [Google Scholar] [CrossRef] [PubMed]
  385. Sun, L.H.; Ban, T.; Liu, C.D.; Chen, Q.X.; Wang, X.; Yan, M.L.; Hu, X.L.; Su, X.L.; Bao, Y.N.; Sun, L.L. Activation of Cdk5/p25 and tau phosphorylation following chronic brain hypoperfusion in rats involves micro RNA-195 down-regulation. J. Neurochem. 2015, 134, 1139–1151. [Google Scholar] [CrossRef] [PubMed]
  386. Ai, J.; Sun, L.-H.; Che, H.; Zhang, R.; Zhang, T.-Z.; Wu, W.-C.; Su, X.-L.; Chen, X.; Yang, G.; Li, K.; et al. MicroRNA-195 Protects Against Dementia Induced by Chronic Brain Hypoperfusion via Its Anti-Amyloidogenic Effect in Rats. J. Neurosci. 2013, 33, 3989–4001. [Google Scholar] [CrossRef] [PubMed]
  387. Tsai, T.H.; Sun, C.K.; Su, C.H.; Sung, P.H.; Chua, S.; Zhen, Y.Y.; Leu, S.; Chang, H.W.; Yang, J.L.; Yip, H.K. Sitagliptin attenuated brain damage and cognitive impairment in mice with chronic cerebral hypo-perfusion through suppressing oxidative stress and inflammatory reaction. J. Hypertens. 2015, 33, 1001–1013. [Google Scholar] [CrossRef]
  388. Flores, G.; Flores-Gomez, G.D.; de Jesus Gomez-Villalobos, M. Neuronal changes after chronic high blood pressure in animal models and its implication for vascular dementia. Synapse 2016, 70, 198–205. [Google Scholar] [CrossRef]
  389. Rohrer, J.D.; Warren, J.D. Phenotypic signatures of genetic frontotemporal dementia. Curr. Opin. Neurol. 2011, 24, 542–549. [Google Scholar] [CrossRef]
  390. Weingarten, M.D.; Lockwood, A.H.; Hwo, S.Y.; Kirschner, M.W. A protein factor essential for microtubule assembly. Proc. Natl. Acad. Sci. USA 1975, 72, 1858–1862. [Google Scholar] [CrossRef] [Green Version]
  391. Drubin, D.G.; Kirschner, M.W. Tau protein function in living cells. J. Cell Biol. 1986, 103, 2739–2746. [Google Scholar] [CrossRef] [Green Version]
  392. Saxena, A.K.; Abdul-Majeed, S.S.; Gurtu, S.; Mohamed, W.M. Investigation of redox status in chronic cerebral hypoperfusion-induced neurodegeneration in rats. Appl. Transl. Genom. 2015, 5, 30–32. [Google Scholar] [CrossRef] [Green Version]
  393. Vazquez-Manrique, R.P.; Farina, F.; Cambon, K.; Dolores Sequedo, M.; Parker, A.J.; Millan, J.M.; Weiss, A.; Deglon, N.; Neri, C. AMPK activation protects from neuronal dysfunction and vulnerability across nematode, cellular and mouse models of Huntington’s disease. Hum. Mol. Genet. 2016, 25, 1043–1058. [Google Scholar] [CrossRef]
  394. Cai, Z.; Yan, L.J.; Li, K.; Quazi, S.H.; Zhao, B. Roles of AMP-activated protein kinase in Alzheimer’s disease. Neuromolecular Med. 2012, 14, 1–14. [Google Scholar] [CrossRef] [PubMed]
  395. Rui, Y.; Myers, K.R.; Yu, K.; Wise, A.; De Blas, A.L.; Hartzell, H.C.; Zheng, J.Q. Activity-dependent regulation of dendritic growth and maintenance by glycogen synthase kinase 3beta. Nat. Commun. 2013, 4, 2628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  396. Zou, C.; Shi, Y.; Ohli, J.; Schuller, U.; Dorostkar, M.M.; Herms, J. Neuroinflammation impairs adaptive structural plasticity of dendritic spines in a preclinical model of Alzheimer’s disease. Acta Neuropathol. 2016, 131, 235–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  397. Szabo, C.; Kelemen, O.; Keri, S. Low-grade inflammation disrupts structural plasticity in the human brain. Neuroscience 2014, 275, 81–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  398. Dahle, C.L.; Jacobs, B.S.; Raz, N. Aging, vascular risk, and cognition: Blood glucose, pulse pressure, and cognitive performance in healthy adults. Psychol. Aging. 2009, 24, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  399. VanGuilder, H.D.; Farley, J.A.; Yan, H.; Van Kirk, C.A.; Mitschelen, M.; Sonntag, W.E.; Freeman, W.M. Hippocampal dysregulation of synaptic plasticity-associated proteins with age-related cognitive decline. Neurobiol. Dis. 2011, 43, 201–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  400. Foster, T.C. Involvement of hippocampal synaptic plasticity in age-related memory decline. Brain Res. Brain Res. Rev. 1999, 30, 236–249. [Google Scholar] [CrossRef]
  401. Small, G.W. What we need to know about age related memory loss. BMJ 2002, 324, 1502–1505. [Google Scholar] [CrossRef] [Green Version]
  402. Temido-Ferreira, M.; Coelho, J.E.; Pousinha, P.A.; Lopes, L.V. Novel Players in the Aging Synapse: Impact on Cognition. J. Caffeine Adenosine Res. 2019, 9, 104–127. [Google Scholar] [CrossRef]
  403. Bettio, L.E.B.; Rajendran, L.; Gil-Mohapel, J. The effects of aging in the hippocampus and cognitive decline. Neurosci. Biobehav. Rev. 2017, 79, 66–86. [Google Scholar] [CrossRef]
  404. Kumar, A. Calcium Signaling During Brain Aging and Its Influence on the Hippocampal Synaptic Plasticity. Adv. Exp. Med. Biol. 2020, 1131, 985–1012. [Google Scholar] [CrossRef]
  405. Dahan, L.; Rampon, C.; Florian, C. Age-related memory decline, dysfunction of the hippocampus and therapeutic opportunities. Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 102, 109943. [Google Scholar] [CrossRef]
  406. Navakkode, S.; Liu, C.; Soong, T.W. Altered function of neuronal L-type calcium channels in ageing and neuroinflammation: Implications in age-related synaptic dysfunction and cognitive decline. Ageing Res. Rev. 2018, 42, 86–99. [Google Scholar] [CrossRef] [PubMed]
  407. Diaz, A.; Trevino, S.; Vazquez-Roque, R.; Venegas, B.; Espinosa, B.; Flores, G.; Fernandez, G.J.; Montano, L.F.; Guevara, J. The aminoestrogen prolame increases recognition memory and hippocampal neuronal spine density in aged mice. Synapse 2017, 71, e21987. [Google Scholar] [CrossRef] [PubMed]
  408. Hatch, R.J.; Leinenga, G.; Gotz, J. Scanning Ultrasound (SUS) Causes No Changes to Neuronal Excitability and Prevents Age-Related Reductions in Hippocampal CA1 Dendritic Structure in Wild-Type Mice. PLoS ONE 2016, 11, e0164278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  409. Markham, J.A.; McKian, K.P.; Stroup, T.S.; Juraska, J.M. Sexually dimorphic aging of dendritic morphology in CA1 of hippocampus. Hippocampus 2005, 15, 97–103. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic flow diagram of the correlation between etiopathogenesis and hippocampal synaptic dysfunction in neurodegenerative diseases. In neurodegenerative diseases, several etiological factors impact hippocampal synaptic plasticity, leading to functional outcomes such as cognition and emotional dysregulations. Abbreviations: Ach, acetylcholine; A2AR, adenosine A2A receptor; AD, Alzheimer’s disease; α-Syn, alpha-synuclein; Aβ, amyloid beta; ALS, amyotrophic lateral sclerosis; BDNF, brain-derived neurotrophic factor; DA, dopamine; EPSC, excitatory post-synaptic current; FTD, frontotemporal dementia; Glu, glutamate; HD, Huntington’s disease; HPtau, hyperphosphorylated Tau; IC, inflammatory cells; IM, inflammatory mediators; LTD, long term depression; LTP, long term potentiation; mhtt, mutant-huntingtin; MS, multiple sclerosis; NA, noradrenaline; NFTs, neurofibrillary tangles; NMDAR, N-methyl-D-aspartate receptors; NTs, neurotransmitter systems; PD, Parkinson’s disease; PSD, post-synaptic density; PTP, post-tetanic potentiation; ROS, reactive oxygen species; VD, vascular dementia.
Figure 1. Schematic flow diagram of the correlation between etiopathogenesis and hippocampal synaptic dysfunction in neurodegenerative diseases. In neurodegenerative diseases, several etiological factors impact hippocampal synaptic plasticity, leading to functional outcomes such as cognition and emotional dysregulations. Abbreviations: Ach, acetylcholine; A2AR, adenosine A2A receptor; AD, Alzheimer’s disease; α-Syn, alpha-synuclein; Aβ, amyloid beta; ALS, amyotrophic lateral sclerosis; BDNF, brain-derived neurotrophic factor; DA, dopamine; EPSC, excitatory post-synaptic current; FTD, frontotemporal dementia; Glu, glutamate; HD, Huntington’s disease; HPtau, hyperphosphorylated Tau; IC, inflammatory cells; IM, inflammatory mediators; LTD, long term depression; LTP, long term potentiation; mhtt, mutant-huntingtin; MS, multiple sclerosis; NA, noradrenaline; NFTs, neurofibrillary tangles; NMDAR, N-methyl-D-aspartate receptors; NTs, neurotransmitter systems; PD, Parkinson’s disease; PSD, post-synaptic density; PTP, post-tetanic potentiation; ROS, reactive oxygen species; VD, vascular dementia.
Ijms 23 03349 g001
Figure 2. Schematic illustration of proposed mechanistic pathways for alteration of structural plasticity in the hippocampus in neurodegenerative diseases. During neurodegenerative diseases, pathological proteins, inflammation, neurotransmitter imbalance, impaired energy production, genetic factors, and activation of kinases affect the actin cytoskeleton and/or microtubule arrangement through different pathways. Abbreviations: AD, Alzheimer’s disease; α-Syn, alpha-synuclein; Aβ, amyloid-β; ATP, Adenosine 5′-triphosphate; BDNF, brain-derived neurotrophic factor; DA, dopamine; FTD, frontotemporal dementia; Glu, glutamate; GluR, glutamate receptor; GSK-3β, glycogen synthase kinase3β; HD, Huntington’s disease; IFNγ, interferon γ; IL-1β, interleukin 1β; LRRK2, leucine-rich repeat kinase 2; LIMK1, LIM kinase 1; MAPT, microtubule-associated tau; MS, multiple sclerosis; NA, noradrenaline; NT, neurotransmitter; PAK1, P21 (RAC1) Activated Kinase 1; PD, Parkinson’s disease; PSD, post-synaptic density; HPtau, hyperphosphorylated Tau; Rac1, Ras-related C3 botulinum toxin substrate 1; TNFα, tumor necrosis factor α; TrkB, tyrosine receptor kinase B; VD, vascular dementia.
Figure 2. Schematic illustration of proposed mechanistic pathways for alteration of structural plasticity in the hippocampus in neurodegenerative diseases. During neurodegenerative diseases, pathological proteins, inflammation, neurotransmitter imbalance, impaired energy production, genetic factors, and activation of kinases affect the actin cytoskeleton and/or microtubule arrangement through different pathways. Abbreviations: AD, Alzheimer’s disease; α-Syn, alpha-synuclein; Aβ, amyloid-β; ATP, Adenosine 5′-triphosphate; BDNF, brain-derived neurotrophic factor; DA, dopamine; FTD, frontotemporal dementia; Glu, glutamate; GluR, glutamate receptor; GSK-3β, glycogen synthase kinase3β; HD, Huntington’s disease; IFNγ, interferon γ; IL-1β, interleukin 1β; LRRK2, leucine-rich repeat kinase 2; LIMK1, LIM kinase 1; MAPT, microtubule-associated tau; MS, multiple sclerosis; NA, noradrenaline; NT, neurotransmitter; PAK1, P21 (RAC1) Activated Kinase 1; PD, Parkinson’s disease; PSD, post-synaptic density; HPtau, hyperphosphorylated Tau; Rac1, Ras-related C3 botulinum toxin substrate 1; TNFα, tumor necrosis factor α; TrkB, tyrosine receptor kinase B; VD, vascular dementia.
Ijms 23 03349 g002
Table 1. Recent evidences of the alteration of structural plasticity in the hippocampus in neurodegenerative diseases.
Table 1. Recent evidences of the alteration of structural plasticity in the hippocampus in neurodegenerative diseases.
Model Dendritic
Complexity
Spine Density/
Morphology
MechanismRef.
ADClinical patientsDecreasedDecreased [279]
APP/PS1-TG miceDecreasedDecreased spine density and mushroom [114]
APP/PS1-TG miceDecreased Fibrillary amyloid deposition[277]
APP/PS1-TG miceDecreased Fibrillary amyloid deposition [277]
APP/PS1-TG mice DecreasedDecreasedAβ plaques pathology[280]
APOE4-TG miceDecreasedDecreased [281]
miR-34c-transfected mouse primary cultured hippocampal neuronsDecreasedDecreased density and
filopodia
[282]
N-tau-TG miceIncreasedIncreased [283]
TgCRND8 miceNo changeDecreased, thin, and stubbyAβ plaques pathology
Alteration of GluR
[112]
2576-TG mice DecreasedAβ plaques pathology
Reactive gliosis
[116]
Aβ-infused rats Decreased [113]
Aβ-infused rats Decreased spine density and length Decreased, thin, and filopodiaGlu circulation[115]
Aβ-treated rat hippocampal slices DecreasedNMDAR[117]
APP/PS1-TG mice Decreased spine length
Increased neck size
[284]
APP-knock-in mice Decreased mushroomDownregulation of synaptic STIM2–nSOC–CaMKII
pathway
[285]
PDLRRK2-mutant mouse primary cultured hippocampal neuronsDecreased LRRK2 regulation[286]
LRRK2-mutant miceDecreased LRRK2 regulation[287]
LRRK2-mutant miceDecreasedDecreasedLRRK2 regulation[288]
MPTP-lesioned mice Decreased [139]
MPTP-lesioned miceNo changeDecreased [289]
PFF-treated primary cultured hippocampal neurons Decreased mushroomα-synucleinopathy[290]
PFF-treated primary cultured hippocampal neurons Decreased spine density and mushroomα-Syn induced dysregulation of the actin-binding protein[291]
HDR6/1-TG miceDecreased [292]
Hip14-deficient mice Decreased [293]
R6/1-TG mice DecreasedNF-κB signaling[294]
Pyk2-deficient mice DecreasedPyk2 regulation[172]
R6/1-TG mice No change [295]
MSEAE miceDecreased [296]
EAE miceDecreased [297]
Cuprizone-diet fed mice No change in total density
Increased mushroom
[298]
VDBCCAO ratsDecreasedDecreased spine density and mushroom [65]
BCCAO ratsDecreasedDecreasedAPP[299]
BCCAO rats Decreased [300]
BCCAO rats Decreased spine density and mushroom [301]
BCCAO rats DecreasedSuppression of AMPK pathway[302]
ALS/FTDTDP-43 overexpressing primary cultured hippocampal neuronsDecreased RNA-binding function of
TDP-43
[67]
ALSSOD1G93A TG miceNo changeNo change [303]
Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; Aβ, amyloid β; APP/PS1, amyloid precursor protein/presenilin 1; AMPK, AMP-activated protein kinase; APOE4, apolipoprotein E4; α-Syn, alpha-synuclein; BCCAO, bilateral common carotid artery occlusion; BDNF, brain-derived neurotrophic factor; CaMKII, calmodulin-dependent protein kinase II; EAE, experimental autoimmune encephalomyelitis; FTD, frontotemporal dementia; Glu, glutamate; GluR, glutamate receptor; HD, Huntington’s disease; LRRK2, leucine-rich repeat kinase 2; miR, microRNA; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MS, multiple sclerosis; NF-κB, nuclear factor kappa B; NMDAR, N-methyl D-aspartate receptor; PD, Parkinson’s disease; PFF- α-Syn preformed fibrils; Pyk2, proline-rich tyrosine kinase 2; STIM2, stromal interaction molecule 2; TDP-43, TAR DNA binding protein-43; TG, transgenic; TgCRND8, transgenic CRND8; VD, vascular dementia.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Weerasinghe-Mudiyanselage, P.D.E.; Ang, M.J.; Kang, S.; Kim, J.-S.; Moon, C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 3349. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23063349

AMA Style

Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim J-S, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2022; 23(6):3349. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23063349

Chicago/Turabian Style

Weerasinghe-Mudiyanselage, Poornima D. E., Mary Jasmin Ang, Sohi Kang, Joong-Sun Kim, and Changjong Moon. 2022. "Structural Plasticity of the Hippocampus in Neurodegenerative Diseases" International Journal of Molecular Sciences 23, no. 6: 3349. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23063349

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