Advertisement
Archival Report|Articles in Press

Compulsive-like behaviors in Aβ1-42 induced Alzheimer’s disease in mice are associated with hippocampo-cortical neural circuit dysfunction

  • Apoorva Bettagere Shivakumar
    Affiliations
    Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
    Search for articles by this author
  • Sparsha Kumari
    Affiliations
    Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
    Search for articles by this author
  • Sonam Fathima Mehak
    Affiliations
    Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
    Search for articles by this author
  • Gireesh Gangadharan
    Correspondence
    Address for correspondence: Gireesh Gangadharan, Ph.D. Assistant Professor/ DBT-Ramalingaswami Fellow, Department of Cell and Molecular biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka 576104, India, OR , Phone: +91-8129066565
    Affiliations
    Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
    Search for articles by this author
Open AccessPublished:March 06, 2023DOI:https://doi.org/10.1016/j.bpsgos.2023.02.009

      ABSTRACT

      BACKGROUND

      In addition to memory deficits, patients with Alzheimer's disease (AD) experience neuropsychiatric disturbances as well. Recent studies have suggested the association of obsessive-compulsive disorder (OCD) with the early stages of AD. However, there is a lack of understanding of the neurobiological underpinnings of compulsive-like behaviors at the neuronal circuit level and their relationship with AD.

      METHODS

      We have addressed this issue in an Aβ1-42 induced mouse model for AD and studied compulsive-like behaviors. Next, we compared the hippocampal and medial prefrontal cortex (mPFC) local field potential (LFP) pattern and coherence between these regions of control and AD mice. We also assessed the expression pattern of acetylcholine and glutamatergic signaling in these regions using quantitative PCR.

      RESULTS

      Here, our findings show that AD mice exhibit compulsive-like behaviors as evidenced by enhanced marble burying, nest building, and burrowing behaviors. Further, AD mice exhibited hippocampo-cortical circuit dysfunction, demonstrated by decreased power of rhythmic oscillations at the theta (4-12Hz) and gamma frequency (25-50Hz) in the hippocampus (HPC) and the mPFC, two functionally interconnected brain regions involved both in AD and compulsive behaviors. Importantly, coherence between the HPC and mPFC in the theta band of AD animals was significantly reduced. Furthermore, we found reduced cholinergic and glutamatergic neurotransmission in the HPC and mPFC of AD mice.

      CONCLUSION

      We conclude that the hippocampo-cortical functional alterations may play a significant role in mediating the compulsive-like behaviors observed in AD mice. These findings may help in understanding the underlying circuit mechanisms of OCD-like phenotypes associated with AD.

      Keywords

      INTRODUCTION:

      Emerging evidence indicates that the neurodegenerative pathology of Alzheimer disease (AD) can lead to neuropsychiatric disturbances at certain stages during the disease progression (
      • Geda YE
      • Roberts RO
      • Knopman DS
      • Petersen RC
      • Christianson TJH
      • Pankratz VS
      • et al.
      Prevalence of Neuropsychiatric Symptoms in Mild Cognitive Impairment and Normal Cognitive Aging: Population-Based Study.
      ,
      • Lanctôt KL
      • Amatniek J
      • Ancoli-Israel S
      • Arnold SE
      • Ballard C
      • Cohen-Mansfield J
      • et al.
      Neuropsychiatric signs and symptoms of Alzheimer’s disease: New treatment paradigms.
      ,
      • Rosenberg PB
      • Nowrangi MA
      • Lyketsos CG
      Neuropsychiatric symptoms in Alzheimer’s disease: What might be associated brain circuits?.
      ). Thus, neuropsychiatric symptoms (NPS) are being increasingly recognized as one of the key features of AD and related dementias. Recent studies have proposed a possible link between AD and obsessive-compulsive disorder (OCD) (
      • Dondu A
      • Sevincoka L
      • Akyol A
      • Tataroglu C
      Is Obsessive–Compulsive symptomatology a risk factor for Alzheimer-type dementia?.
      ,
      • Mrabet Khiari H
      • Achouri A
      • Ben Ali N
      • Cherif A
      • Batti H
      • Messaoud T
      • Mrabet A
      Obsessive-compulsive disorder: a new risk factor for Alzheimer disease?.
      ,
      • Ruggeri M
      • Ricci M
      • Gerace C
      • Blundo C
      Late-onset obsessive-compulsive disorder as the initial manifestation of possible behavioural variant Alzheimer’s disease.
      ), a common disabling neuropsychiatric disorder characterized by constant intrusive thoughts and compulsive behaviors (
      • Robbins TW
      • Vaghi MM
      • Banca P
      Obsessive-Compulsive Disorder: Puzzles and Prospects.
      ). However, the interaction between OCD and AD pathology is poorly understood. Structural and functional neuroimaging studies suggest that a wide range of brain regions and circuits, in particular the cortico-striato-thalamo-cortical circuit (CSTC), are implicated in the pathophysiology of OCD (
      • Graybiel AM
      • Rauch SL
      Toward a Neurobiology of Obsessive-Compulsive Disorder.
      ,
      • Milad MR
      • Rauch SL
      Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
      ). These are also demonstrated in animal studies, for example, Sapap3- and Slitrk5-mutant mouse models of OCD exhibited defects in cortico-striatal synapses and OCD-like phenotypes (
      • Shmelkov SV
      • Hormigo A
      • Jing D
      • Proenca CC
      • Bath KG
      • Milde T
      • et al.
      Slitrk5 deficiency impairs corticostriatal circuitry and leads to obsessive-compulsive–like behaviors in mice [no. 5].
      ,
      • Welch JM
      • Lu J
      • Rodriguiz RM
      • Trotta NC
      • Peca J
      • Ding J-D
      • et al.
      Cortico-striatal synaptic defects and OCD-like behaviours in Sapap3-mutant mice.
      ).
      Intriguingly, structural imaging in humans (
      • Milad MR
      • Rauch SL
      Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
      ,
      • Boedhoe PSW
      • Schmaal L
      • Abe Y
      • Ameis SH
      • Arnold PD
      • Batistuzzo MC
      • et al.
      Distinct Subcortical Volume Alterations in Pediatric and Adult OCD: A Worldwide Meta- and Mega-Analysis.
      ,
      • Menzies L
      • Chamberlain SR
      • Laird AR
      • Thelen SM
      • Sahakian BJ
      • Bullmore ET
      Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
      ,
      • Rao S
      • Raveendranathan D
      • Shivakumar V
      • Narayanaswamy JC
      • Venkatasubramanian G
      • Reddy YCJ
      Hippocampus volume alterations and the clinical correlates in medication naïve obsessive compulsive disorder.
      ,
      • Reess TJ
      • Rus OG
      • Gürsel DA
      • Schmitz-Koep B
      • Wagner G
      • Berberich G
      • Koch K
      Association between hippocampus volume and symptom profiles in obsessive–compulsive disorder.
      ,
      • Schmitz TW
      • Correia MM
      • Ferreira CS
      • Prescot AP
      • Anderson MC
      Hippocampal GABA enables inhibitory control over unwanted thoughts [no. 1].
      ,
      • Ullrich M
      • Weber M
      • Post AM
      • Popp S
      • Grein J
      • Zechner M
      • et al.
      OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency.
      ) and lesion studies in rodents (
      • Deacon RMJ
      • Croucher A
      • Rawlins JNP
      Hippocampal cytotoxic lesion effects on species-typical behaviours in mice.
      ,
      • Deacon RMJ
      • Penny C
      • Rawlins JNP
      Effects of medial prefrontal cortex cytotoxic lesions in mice.
      ) have suggested that the hippocampus (HPC) and medial prefrontal cortex (mPFC) play an essential role in compulsive behaviors. A recent study reported that the functional connectivity between HPC and mPFC is important for the regulation of invasive thoughts (
      • Schmitz TW
      • Correia MM
      • Ferreira CS
      • Prescot AP
      • Anderson MC
      Hippocampal GABA enables inhibitory control over unwanted thoughts [no. 1].
      ). The interaction between these two regions is regulated by neuronal oscillations, especially rhythmic oscillations at the theta frequency (4-12Hz) during behaviors (
      • Colgin LL
      Oscillations and hippocampal–prefrontal synchrony.
      ,
      • O’Neill P-K
      • Gordon JA
      • Sigurdsson T
      Theta Oscillations in the Medial Prefrontal Cortex Are Modulated by Spatial Working Memory and Synchronize with the Hippocampus through Its Ventral Subregion.
      ). Several neurotransmitters, including acetylcholine, are known to regulate the fine-tuning of hippocampo-medial prefrontal cortex (HPC-mPFC) functional integration (
      • Ruggiero RN
      • Rossignoli MT
      • Marques DB
      • de Sousa BM
      • Romcy-Pereira RN
      • Lopes-Aguiar C
      • Leite JP
      Neuromodulation of Hippocampal-Prefrontal Cortical Synaptic Plasticity and Functional Connectivity: Implications for Neuropsychiatric Disorders.
      ). Although dysfunction of neurotransmission in the CSTC circuits, including serotonin, dopamine, and glutamate systems are reported in the OCD condition (
      • Sun T
      • Song Z
      • Tian Y
      • Tian W
      • Zhu C
      • Ji G
      • et al.
      Basolateral amygdala input to the medial prefrontal cortex controls obsessive-compulsive disorder-like checking behavior.
      ), recent evidences point toward the involvement of central cholinergic systems in OCD (
      • Dondu A
      • Sevincoka L
      • Akyol A
      • Tataroglu C
      Is Obsessive–Compulsive symptomatology a risk factor for Alzheimer-type dementia?.
      ).
      These previous findings present a possibility that hippocampo-cortical function may be critically associated with compulsive behaviors in AD. We have tested this possibility by assessing and observing compulsive behavioral phenotype in Aβ1-42 induced AD mice. Furthermore, theta and gamma oscillations in the HPC and the mPFC were reduced in AD mice. Additionally, the coherence at the theta frequency oscillations between the HPC and mPFC neuronal circuit was disrupted in AD mice. In support of this, we found downregulation of acetylcholine and glutamatergic signaling in the hippocampo-cortical neuronal pathway. These results suggest that hippocampo-cortical neural circuit may play a significant role in the compulsive behaviors associated with AD.

      MATERIALS AND METHODS

      Animals

      C57BL/6 male mice, aged 8-10-week-old and weighing 25-30g were used in this study. Animal handling and care was done in adherence to the Institutional animal ethical committee (IAEC), Kasturba Medical College (KMC), Manipal Academy of Higher Education (MAHE), Manipal, India (IAEC/KMC/05/ 2021, dated 23/01/2021). Briefly, four animals were housed per cage at a temperature of 22±2°C under a 12:12h light-dark schedule with light beginning at 6:30 A.M. Food and water were provided ad libitum.

      Alzheimer’s disease induction in mice

      Amyloid β Protein Fragment1-42 (A9810, Sigma, USA) was procured and Aβ1-42 oligomers (AβO) were prepared and injections were performed as previously described (

      Stine WB, Jungbauer L, Yu C, LaDu MJ (2010): Preparing Synthetic Aβ in Different Aggregation States. In: Roberson ED, editor. Alzheimer’s Disease and Frontotemporal Dementia, vol. 670. Totowa, NJ: Humana Press, pp 13–32.

      ,
      • Wang L-S
      • Tao X
      • Liu X-M
      • Zhou Y-F
      • Zhang M-D
      • Liao Y-H
      • et al.
      Cajaninstilbene Acid Ameliorates Cognitive Impairment Induced by Intrahippocampal Injection of Amyloid-β1–42 Oligomers.
      ). A detailed description is provided in the Supplement.

      Behavioral assays

      All behavioral tests were performed during the daytime (9 AM to 5 PM). The mice were subjected to the following experiments: compulsive-like behavior assays (marble burying, nesting and, burrowing), spatial memory test, hole board assay, open field test, elevated plus maze, light-dark and forced swim test. Behavioral analysis was performed using the behavioral tracking system EthoVision XT 15 (Noldus, Netherlands). A detailed description of behavioural tests is provided in the Supplement.

      In Vivo Local Field-Potential (LFP) Recordings and Data Analysis:

      Hippocampal and mPFC LFP were recorded from control and AD mice using published protocols (
      • Gangadharan G
      • Shin J
      • Kim S-W
      • Kim A
      • Paydar A
      • Kim D-S
      • et al.
      Medial septal GABAergic projection neurons promote object exploration behavior and type 2 theta rhythm.
      ,
      • Shin J
      • Gireesh G
      • Kim S-W
      • Kim D-S
      • Lee S
      • Kim Y-S
      • et al.
      Phospholipase C 4 in the Medial Septum Controls Cholinergic Theta Oscillations and Anxiety Behaviors.
      ). Briefly, for the electrode implantation, mice were anesthetized as mentioned earlier and positioned on a stereotaxic apparatus and bregma and lambda oriented to be in the same horizontal surface. A Teflon-coated tungsten electrode (Cat No#796500, A-M Systems, USA) was unilaterally implanted into the hippocampal fissure (from bregma, AP -2.0 mm, ML ±1.2 mm, and DV -1.8 mm) and mPFC (from bregma, AP +1.7 mm, ML ±0.3 mm, and DV -2.0 mm) on the right hemisphere for LFP recording with grounding over the cerebellum. The reference electrode was implanted in the frontal lobe. LFP signals were amplified and digitized at a sampling frequency of 1,000 Hz. Data were acquired using an RHS stim/recording system (Intan Technologies, USA). The recording placements for hippocampal and mPFC LFP were verified by post-mortem histology (Nissl-staining) (Supplementary Figure S1).
      The LFP data were analyzed offline in MATLAB (R2021a, MathWorks, Natick MA, USA) by the means of built-in and custom-written routines. The Welch periodogram technique was used to determine power spectral density using the pwelch.m function from the Signal Processing Toolbox (50% overlapping, 4 s Hamming windows), and the mean spectral power was calculated from artifact-free one-minute LFP recordings made from each animal. Time-frequency power decomposition was achieved using the spectrogram.m function. The coherence between the hippocampal and mPFC LFP was measured by magnitude-squared coherence using the mscohere.m function.

      Immunohistochemistry and Real-time quantitative PCR

      A detailed description of immunohistochemistry and real-time quantitative PCR is provided in the Supplement.

      Statistics

      Results are illustrated as mean ± SEM. Data were evaluated by the student’s t-test, with a significance level achieved when P<0.05, using Sigmaplot software (Systat Software).

      RESULTS

      AD mice exhibited compulsive-like behavior

      As a first step, we aimed at characterizing the compulsive-like behaviors in Aβ1-42 induced AD mice. We tested the AD and control mice in three behavioral assays that measure stereotyped compulsive-like behavior in rodents: marble burying, nestlet shredding, and burrowing tests. In the marble burying test, AD mice buried a significantly greater number of marbles compared to the control group (Figure 1B, C) (Marbles buried, control: 10.50 ± 1.11, AD: 17.50 ± 0.71; p=0.0006, t=5.26, student’s t-test). In the nestlet shredding test, the AD mice demonstrated better performance in shredding and accomplished higher nest quality scores than the control mice (Figure 1D, E) (Nest ratings, control: 2.83 ± 0.60, AD: 4.66 ± 0.21; p=0.02, t=2.87, student’s t-test). Similarly, in the burrowing test, a significantly larger amount of food pellets was burrowed out of the tunnel by AD mice in comparison to the control (Figure 1F, G) (% of burrowed material, control: 12.76 ± 5.94, AD: 88.77 ± 6.96, p<0.0001, t=8.30, student’s t-test). Together, these findings demonstrated and confirmed compulsive-like behaviors in AD mice.
      Figure thumbnail gr1
      Figure 1(A-G) AD mice exhibited compulsive-like behavior. (A) Illustration of intra-hippocampal injection of amyloid beta 1-42 (B) Representative images of the marble burying test (red arrows for 2/3 buried after test) (C) AD mice displayed a greater number of marbles buried (D) Representative images of the nest building test (E) AD mice built nests of better quality (F) Representative images of the burrowing test (G) AD mice burrowed a higher percentage of food pellets. (H-I) AD mice displayed decreased novelty-seeking behavior (H) Number of head dips (I) time spent in head dipping during the hole board test. (J) AD mice displayed depressive like behavior. AD mice remained immobile for a longer duration. Values represent mean ± SEM. Control n=6, AD n=6; *P < 0.05, **P < 0.01 and ***P < 0.001.

      Reduced novelty-seeking behavior in AD mice

      It is known that novelty-seeking behavior, a fundamental trait that drives exploratory behavior, is reduced in individuals with OCD (
      • Kusunoki K
      • Sato T
      • Taga C
      • Yoshida Y
      • Komori K
      • Narita T
      • et al.
      Low novelty-seeking differentiates obsessive-compulsive disorder from major depression: Low novelty-seeking in OCD.
      ). The head-dipping measure in the hole-board test is generally considered a valid measure of novelty-seeking behavior (
      • Abreu-Villaça Y
      • Queiroz-Gomes F do E
      • Dal Monte AP
      • Filgueiras CC
      • Manhães AC
      Individual differences in novelty-seeking behavior but not in anxiety response to a new environment can predict nicotine consumption in adolescent C57BL/6 mice.
      ). The AD mice displayed a significant reduction in the number and duration of head dips (Figure 1H) (number of head dips, control: 21.00 ± 1.26, AD: 9.33 ± 1.20, p<0.0001, t=6.68; time spent dipping (Figure 1I), control: 10.64 ± 0.93, AD: 5.64 ± 0.53, p=0.001, t=4.62, student’s t-test). These findings suggested decreased novelty-seeking behavior in AD mice.

      AD mice displayed depressive-like behavior

      OCD is frequently accompanied by multiple disorders, with depression being the most common comorbidity (
      • Abramowitz JS
      • Storch EA
      • Keeley M
      • Cordell E
      Obsessive-compulsive disorder with comorbid major depression: What is the role of cognitive factors?.
      ,
      • Tibi L
      • van Oppen P
      • van Balkom AJLM
      • Eikelenboom M
      • Rickelt J
      • Schruers KRJ
      • Anholt GE
      The long-term association of OCD and depression and its moderators: A four-year follow up study in a large clinical sample.
      ). To evaluate depression-like symptoms in AD mice, we performed the forced swim test, one of the most widely used behavioral paradigm for the investigation of depressive-like state in rodents. The FST draws on the notion that an animal placed in a container filled with water would struggle to escape at first, but gradually become immobile, which might be read as a symptom of behavioural despair. AD mice spent significantly less time in the tank trying to escape (mobility, climbing, and swimming) (Figure 1J) in comparison with the control group (mobile time, control: 92.33 ± 14.15, AD: 50.83 ± 6.26, p=0.03, t=2.68, student’s t-test). These results indicated depressive-like behavior in AD mice.

      Unchanged anxiety levels in AD mice

      The light-dark transition test and EPM were conducted to evaluate whether anxiety was comorbid with compulsive-like behaviors. The transition from dark to light (Figure 2A) (control: 7.66 ± 1.25, AD: 8.00 ± 1.46, p=0.86, t=0.17) and the total time spent in the light chamber (Figure 2B) (control: 96.16 ± 24.09, AD: 103.50±23.04, p=0.83, t=0.22, student’s t-test) remained similar between the AD and control groups. Similarly, no significant difference in the percentage of open arm entries (Figure 2C) (control: 40.40 ±3.59, AD: 42.35 ±3.79; p=0.71, t=0.37), total entries (Figure 2D) (control: 19.50 ±2.10, AD: 17.66 ±2.51; p=0.58, t=0.55), and total distance travelled (Figure 2E) (control: 1171.70 ±80.34, AD: 985.42 ±114.27; p=0.21, t=1.33, student’s t-test) were observed in EPM assay. These results indicated that Aβ1-42 induced AD mice do not exhibit anxiety-like behavior.
      Figure thumbnail gr2
      Figure 2AD mice displayed (A-E) intact anxiety like behavior in (A-B) light-dark, (C-E) elevated plus maze test and (F-H) locomotor activity in open field test. (A) Number of transitions to light compartment (B) Time spent in light compartment. (C) Percentage of open arm entries and (D) Total number of entries (E) Total distance travelled (F) Number of times mice entered the center of the open field. (G) Distance travelled across 5 min time bins in the 60 min (H) Total distance travelled in the open field during 60 min window revealing no significant alterations in the locomotor activity. Values represent mean ± SEM. Control n=6, AD n=6; n.s., not significant.

      Intact locomotor activity in AD mice

      To rule out the possibility of any potential locomotion-related abnormalities interfering with the behavioral assays, we performed the open field test for a period of 1 hour and found no difference in the distance traveled and the number of entries into the center between the groups (Figure 2F-H) (frequency of entries, control: 47.00 ± 17.94, AD: 61.83 ± 16.02, p=0.55, t=0.61, total distance moved, control: 8186.40 ± 1230.67 cm, AD: 9617.07 ± 987.54 cm, p=0.38, t=0.90, student’s t-test).

      Impaired spatial memory in AD mice

      The early clinical indication of AD is loss of spatial memory (
      • Pearson-Leary J
      • McNay EC
      Intrahippocampal Administration of Amyloid-β 1-42 Oligomers Acutely Impairs Spatial Working Memory, Insulin Signaling, and Hippocampal Metabolism.
      ). Patients with OCD are also reported to have memory problems (
      • van der Wee NJA
      • Ramsey NF
      • Jansma JM
      • Denys DA
      • van Megen HJGM
      • Westenberg HMG
      • Kahn RS
      Spatial working memory deficits in obsessive compulsive disorder are associated with excessive engagement of the medial frontal cortex.
      ). To confirm any memory deficits in AD mice, we performed the object location recognition test. During the familiarization phase, both control and AD mice exhibited similar preference percentages towards both objects (Figure 3A). In the test phase, the control mice were able to recognize the displaced object and showed more preference for the displaced object. On the other hand, AD mice failed to distinguish between the two objects and showed reduced preference for the displaced object (Figure 3B) (control: 57.21± 0.47; AD: 47.40±1.71, p=0.001, t=5.49, student’s t-test). This data suggested that intrahippocampal injections of Aβ1-42 impaired spatial memory.
      Figure thumbnail gr3
      Figure 3(A-B) AD mice exhibited impaired spatial recognition memory (A) Control and AD mice showed similar preference toward object 1 and object 2 in the familiarization phase (B) AD mice showed significant impairments in remembering the original location of the displaced object. Values represent mean ± SEM. Control n=6, AD n=6; **P < 0.01.

      Reduced LFP oscillation in HPC and mPFC of AD mice

      Rhythmic oscillations of neuronal networks are highly correlated with various cognitive processes (
      • Mehak SF
      • Shivakumar AB
      • Kumari S
      • Muralidharan B
      • Gangadharan G
      Theta and gamma oscillatory dynamics in mouse models of Alzheimer’s disease: A path to prospective therapeutic intervention.
      ). Alterations in brain rhythms have been linked to a variety of neuropsychiatric conditions, including OCD (
      • Lei H
      • Lai J
      • Sun X
      • Xu Q
      • Feng G
      Lateral orbitofrontal dysfunction in the Sapap3 knockout mouse model of obsessive–compulsive disorder.
      ,
      • Pogarell O
      • Juckel G
      • Mavrogiorgou P
      • Mulert C
      • Folkerts M
      • Hauke W
      • et al.
      Symptom-specific EEG power correlations in patients with obsessive–compulsive disorder.
      ). Although there is a consensus on the role of cortico-striatal circuitry in the pathophysiology of obsessive-compulsive disorder (
      • Menzies L
      • Chamberlain SR
      • Laird AR
      • Thelen SM
      • Sahakian BJ
      • Bullmore ET
      Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
      ,
      • Ahmari SE
      • Dougherty DD
      Dissecting Ocd Circuits: From Animal Models to Targeted Treatments.
      ,
      • Fettes P
      • Schulze L
      • Downar J
      Cortico-Striatal-Thalamic Loop Circuits of the Orbitofrontal Cortex: Promising Therapeutic Targets in Psychiatric Illness.
      ), recent evidences have pointed to the key role of the HPC, the primary target region in the AD condition and in the pathology of OCD (
      • Milad MR
      • Rauch SL
      Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
      ,
      • Menzies L
      • Chamberlain SR
      • Laird AR
      • Thelen SM
      • Sahakian BJ
      • Bullmore ET
      Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
      ,
      • Reess TJ
      • Rus OG
      • Gürsel DA
      • Schmitz-Koep B
      • Wagner G
      • Berberich G
      • Koch K
      Association between hippocampus volume and symptom profiles in obsessive–compulsive disorder.
      ,
      • Ullrich M
      • Weber M
      • Post AM
      • Popp S
      • Grein J
      • Zechner M
      • et al.
      OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency.
      ,
      • Wood J
      • Ahmari SE
      A Framework for Understanding the Emerging Role of Corticolimbic-Ventral Striatal Networks in OCD-Associated Repetitive Behaviors.
      ). Thus, understanding functional changes in the hippocampo-cortical neural circuit of AD mice could help to identify potential circuit mechanisms for compulsive behaviors associated with AD. Therefore, we recorded hippocampal LFP to assess any rhythmic alterations that could be present in AD mice. In the power spectral analysis, AD mice demonstrated a significant reduction in the hippocampal theta power (4 – 12 Hz) compared to the control group. However, the gamma (25 – 50 Hz) frequency band in the HPC did not show a statistically significant difference even though the power of gamma was reduced in AD mice (Figure 4D) (Theta power, control: 6.10⨯10-6 ±9.32⨯10-7, AD: 2.82⨯10-6 ±4.45⨯10-7; p=0.009, t=3.17; gamma power, control: 1.77⨯10-6 ±6.01⨯10-7, AD: 7.04⨯10-7 ±1.25⨯10-7; p=0.11, t=1.73, student’s t-test). The mPFC power spectral analysis of the AD mice showed significant reduction in both theta and gamma frequencies compared to the control group (Figure 4E) (Theta power, control: 9.98⨯10-7 ±1.45⨯10-7, AD: 5.81⨯10-7 ±7.05⨯10-8; p=0.02, t=2.58; gamma power, control: 2.66⨯10-7 ±3.80⨯10-8, AD: 1.04⨯10-7 ±9.50⨯10-9; p=0.002, t=4.13, student’s t-test). We also examined the coherence between the HPC and mPFC and observed a significantly reduced coherence in the theta band of the AD animals (Figure 4F) (Theta coherence, control: 0.65 ±0.02, AD: 0.55 ±0.01; p=0.01, t=3.09; gamma coherence control: 0.16 ±0.007, AD: 0.21 ±0.03; p=0.15, t=1.52, student’s t-test). Taken together, these data suggested that the functional interaction between HPC and mPFC is reduced in Aβ1-42 induced AD mice.
      Figure thumbnail gr4
      Figure 4(A-E) Attenuated theta and gamma band oscillations in the HPC and mPFC of AD mice. (A) Illustrate showing in vivo LFP recordings in the HPC and mPFC. (B) Representative oscillatory traces in the HPC and mPFC of control and AD mice under awake state. (C) Representative power spectrogram of control and AD mice (D-E) The oscillatory power at θ and γ band was reduced in (D) HPC and (E) mPFC of AD mice. (F) Impaired theta coherence between the HPC and mPFC of AD mice. (n = 6 control, n = 6 AD). Values represent mean ± SEM. Control n=6, AD n=6; *P < 0.05, **P < 0.01 and n.s., not significant.

      Reduced cholinergic and glutamatergic neurotransmission in the HPC and mPFC of AD mice

      The disrupted LFP oscillations found in the HPC and mPFC of AD mice may indicate dysfunction of the hippocampo-cortical neural circuit. Cholinergic deficits are suggested as a significant contributor to the neuropsychiatric aspect of AD (
      • Cummings JL
      • Back C
      The Cholinergic Hypothesis of Neuropsychiatric Symptoms in Alzheimer’s Disease.
      ,
      • Lanari A
      • Amenta F
      • Silvestrelli G
      • Tomassoni D
      • Parnetti L
      Neurotransmitter deficits in behavioural and psychological symptoms of Alzheimer’s disease.
      ). Interestingly, evidence for cholinergic involvement in compulsive disorder exists (
      • Lucey JV
      • Butcher G
      • Clare AW
      • Dinan TG
      Elevated growth hormone responses to pyridostigmine in obsessive-compulsive disorder: Evidence of cholinergic supersensitivity.
      ,
      • Mitra S
      • Mucha M
      • Khatri SN
      • Glenon R
      • Schulte MK
      • Bult-Ito A
      Attenuation of Compulsive-Like Behavior Through Positive Allosteric Modulation of α4β2 Nicotinic Acetylcholine Receptors in Non-Induced Compulsive-Like Mice.
      ,
      • Piacentino D
      • Maraone A
      • Roselli V
      • Berardelli I
      • Biondi M
      • Kotzalidis GD
      • Pasquini M
      Efficacy of nicotine administration on obsessions and compulsions in OCD: a systematic review.
      ). Additionally, the glutamatergic system has been directly related to the etiology of AD (
      • Yeung JHY
      • Calvo-Flores Guzmán B
      • Palpagama TH
      • Ethiraj J
      • Zhai Y
      • Tate WP
      • et al.
      Amyloid-beta1–42 induced glutamatergic receptor and transporter expression changes in the mouse hippocampus.
      ). Changes in glutamatergic neurotransmission is part of the pathogenesis of OCD (
      • Tsutsui-Kimura I
      • Ohmura Y
      • Izumi T
      • Yamaguchi T
      • Yoshida T
      • Yoshioka M
      Endogenous acetylcholine modulates impulsive action via α4β2 nicotinic acetylcholine receptors in rats.
      ). This evidence prompted us to examine whether the muscarinic/nicotinic cholinergic and glutamatergic neurotransmission in the HPC and mPFC of AD mice were altered. Using real-time PCR, we examined the expression patterns of the genes for muscarinic (CHRM1, CHRM3, CHRM5), nicotinic (CHRNA4, CHRNA7), PLCβ1, and glutamate (GRIN2B) in the dorsal hippocampus (dHPC), ventral hippocampus (vHPC) and medial prefrontal cortex (mPFC) of control and AD mice. The results revealed a statistically significant reduction in the expression patterns of all the genes except CHRM5 gene in the dHPC and mPFC, and CHRNA4 in vHPC (Figure 5) (dHPC: CHRM1: 0.55 ± 0.04, p=0.002, t=9.54; CHRM3: 0.56 ± 0.07, p=0.009, t=5.95; CHRM5: 0.60 ± 0.13, p=0.06, t=2.89; CHRNA4: 0.61 ± 0.11, p=0.04, t=3.27; CHRNA7: 0.53 ± 0.03, p=0.001, t=11.66; PLCβ1: 0.73 ± 0.04, p=0.009, t=5.96; GRIN2B: 0.50 ± 0.12, p=0.02, t=4.08; vHPC: CHRM1: 0.47 ± 0.07, p=0.006, t=6.76; CHRM3: 0.45 ± 0.06, p=0.004, t=7.75; CHRM5: 0.35 ± 0.07, p=0.03, t=8.73; CHRNA4: 0.68 ± 0.24, p=0.29, t=1.27; CHRNA7: 0.47 ± 0.07, p=0.006, t=6.91; PLCβ1: 0.60 ± 0.06, p=0.007, t=6.40; GRIN2B: 0.58 ± 0.03, p=0.001, t=11.66; mPFC: CHRM1: 0.71 ± 0.08, p=0.04, t=3.36; CHRM3: 0.13 ± 0.03, p=0.0002, t=23.48; CHRM5: 0.90 ± 0.13, p=0.53, t=0.69; CHRNA4: 0.39 ± 0.06, p=0.002, t=9.27; CHRNA7: 0.40 ± 0.04, p=0.001, t=12.17; PLCβ1: 0.75 ± 0.07, p=0.04, t=3.39; GRIN2B: 0.69 ± 0.05, p=0.01, t=5.54, student’s t-test). Overall, our molecular experiments revealed that acetylcholine and glutamatergic signalling pathways are altered in the HPC-mPFC neuronal circuit in Aβ1-42 induced AD mice.
      Figure thumbnail gr5
      Figure 5(A-M) Reduced acetylcholine and glutamatergic neurotransmission in the HPC and mPFC of AD mice (A) Pictorial representation of the brain regions used for the quantitative PCR analysis. AD mice showed downregulation of muscarinic (M1, M3, M5), nicotinic (α4, α7), PLCβ1, and glutamate (NR2B) receptor gene expression in the (B-E) dHPC, (F-I) vHPC and (J-M) mPFC. Values represent mean ± SEM. Control n=5, AD n=4; *P < 0.05, **P < 0.01, ***P < 0.001 and n.s., not significant.

      Aβ deposition in the hippocampus of AD mice

      The mice were sacrificed on the tenth day after the last injection to assess amyloid deposition. Hippocampus sections were immunostained for 6E10 positive plaques. Confocal imaging of the immunostained sections revealed clear and widespread depositions of amyloid beta plaques in the hippocampus of AD mouse brain (Figure 6). These results suggest that intra-hippocampal injections of oligomeric assemblies of Aβ1-42 were able to induce amyloid-beta deposition.
      Figure thumbnail gr6
      Figure 6(A-D) Immunostaining of amyloid-β protein deposition in the mouse hippocampus 10 days after the injection of Aβ1-42. Representative HPC sections of Aβ1-42 or vehicle injected mice stained by 6E10 antibody (A-B) Lower magnification (10x) Scale bar, 250 μm and (C-D) higher magnification (63x) Scale bar, 25 μm.

      DISCUSSION

      Compulsivity is an important behavioral aspect that needs to be addressed across disorders and is increasingly becoming a topic of research (
      • Luigjes J
      • Segrave R
      • de Joode N
      • Figee M
      • Denys D
      Efficacy of Invasive and Non-Invasive Brain Modulation Interventions for Addiction.
      ). Several studies have pointed to compulsive behaviors in patients with AD (
      • Mrabet Khiari H
      • Achouri A
      • Ben Ali N
      • Cherif A
      • Batti H
      • Messaoud T
      • Mrabet A
      Obsessive-compulsive disorder: a new risk factor for Alzheimer disease?.
      ,
      • Frydman I
      • Ferreira-Garcia R
      • Borges MC
      • Velakoulis D
      • Walterfang M
      • Fontenelle LF
      Dementia Developing in Late-onset and Treatment-refractory Obsessive-compulsive Disorder.
      ). The current study revealed that Aβ1-42 induced AD mice exhibit a compulsive-like phenotype coupled with a depressive-like state, and also show impaired spatial memory. Additionally, deficits in novelty-seeking, but not anxiogenic-like, behaviors were observed. These behavioral outcomes were accompanied by impaired rhythmic oscillations along with dowregulated expression of acetylcholine and glutamatergic neurotransmission in the HPC-mPFC neuronal circuit. This study is the first to reveal a potential link between the dysfunction of the HPC-mPFC neuronal circuit and compulsive behavior in an AD mouse model.
      AD pathology is accompanied by multiple neurocognitive symptoms during the course of disease progression, and is also known to share similarities with OCD pathology (
      • Dondu A
      • Sevincoka L
      • Akyol A
      • Tataroglu C
      Is Obsessive–Compulsive symptomatology a risk factor for Alzheimer-type dementia?.
      ,
      • Frydman I
      • Ferreira-Garcia R
      • Borges MC
      • Velakoulis D
      • Walterfang M
      • Fontenelle LF
      Dementia Developing in Late-onset and Treatment-refractory Obsessive-compulsive Disorder.
      ). Clinical evidence suggests patients with OCD frequently experience memory disturbances, which are a common symptom of AD patients (
      • Chamberlain SR
      • Fineberg NA
      • Blackwell AD
      • Clark L
      • Robbins TW
      • Sahakian BJ
      A neuropsychological comparison of obsessive–compulsive disorder and trichotillomania.
      ,
      • Purcell R
      • Maruff P
      • Kyrios M
      • Pantelis C
      Cognitive Deficits in Obsessive–Compulsive Disorder on Tests of Frontal–Striatal Function.
      ). We have confirmed that AD mice exhibit impaired spatial memory which is consistent with previous reports (
      • Kasza Á
      • Penke B
      • Frank Z
      • Bozsó Z
      • Szegedi V
      • Hunya Á
      • et al.
      Studies for Improving a Rat Model of Alzheimer’s Disease: Icv Administration of Well-Characterized β-Amyloid 1-42 Oligomers Induce Dysfunction in Spatial Memory [no. 11].
      ,
      • Zhu H
      • Yan H
      • Tang N
      • Li X
      • Pang P
      • Li H
      • et al.
      Impairments of spatial memory in an Alzheimer’s disease model via degeneration of hippocampal cholinergic synapses [no. 1].
      ). In this line, anomalies in the HPC, one of the earliest affected brain regions in AD, as well as the mPFC (
      • Deacon RMJ
      • Croucher A
      • Rawlins JNP
      Hippocampal cytotoxic lesion effects on species-typical behaviours in mice.
      ,
      • Deacon RMJ
      • Penny C
      • Rawlins JNP
      Effects of medial prefrontal cortex cytotoxic lesions in mice.
      ), are suggested to be responsible for compulsive-like behaviors . By HPC specific injection of β-amyloid, we demonstrated that compulsive behaviors are linked to the pathology of AD. Recently, there has been evidence of compulsive behavior in multiple animal models of AD (
      • Filali M
      • Lalonde R
      • Rivest S
      Cognitive and non-cognitive behaviors in an APPswe/PS1 bigenic model of Alzheimer’s disease.
      ,
      • Gimenez-Llort L
      • Alveal-Mellado D
      Digging Signatures in 13-Month-Old 3xTg-AD Mice for Alzheimer’s Disease and Its Disruption by Isolation Despite Social Life Since They Were Born.
      ,
      • Peng Y
      • Cai P
      • Hu J
      • Jiang J
      • Zhang J
      • Liu K
      • et al.
      Altered corticostriatal synchronization associated with compulsive-like behavior in APP/PS1 mice.
      ,
      • Shepherd A
      • May C
      • Churilov L
      • Adlard PA
      • Hannan AJ
      • Burrows EL
      Evaluation of attention in APP/PS1 mice shows impulsive and compulsive behaviours.
      ,
      • Torres-Lista V
      • López-Pousa S
      • Giménez-Llort L
      Marble-burying is enhanced in 3xTg-AD mice, can be reversed by risperidone and it is modulable by handling.
      ,
      • Torres-Lista V
      • Giménez-Llort L
      Impairment of nesting behaviour in 3xTg-AD mice.
      ). For example, enhanced marble-burying behavior is reported in 12-month-old 3xTg-AD male mice (
      • Gimenez-Llort L
      • Alveal-Mellado D
      Digging Signatures in 13-Month-Old 3xTg-AD Mice for Alzheimer’s Disease and Its Disruption by Isolation Despite Social Life Since They Were Born.
      ,
      • Torres-Lista V
      • López-Pousa S
      • Giménez-Llort L
      Marble-burying is enhanced in 3xTg-AD mice, can be reversed by risperidone and it is modulable by handling.
      ) and APP/PS1 AD mice (
      • Peng Y
      • Cai P
      • Hu J
      • Jiang J
      • Zhang J
      • Liu K
      • et al.
      Altered corticostriatal synchronization associated with compulsive-like behavior in APP/PS1 mice.
      ), and elevated nesting behavior in 3xTg AD mice (
      • Torres-Lista V
      • Giménez-Llort L
      Impairment of nesting behaviour in 3xTg-AD mice.
      ). In accordance with this, the current study shows an increase in marble-burying, nest-building, and burrowing behaviors. These findings are suggestive of the use of compulsive behaviors in AD mice as an additional tool for translational assessments of neurocognitive behaviors. It might also be useful in assessing potential risk factors for the disease. Moreover, we noticed reduced novelty-seeking behavior in AD mice. It is known that novelty-seeking behavior is impaired in individuals with OCD (
      • Kusunoki K
      • Sato T
      • Taga C
      • Yoshida Y
      • Komori K
      • Narita T
      • et al.
      Low novelty-seeking differentiates obsessive-compulsive disorder from major depression: Low novelty-seeking in OCD.
      ,
      • Alonso P
      • Menchón JM
      • Jiménez S
      • Segalàs J
      • Mataix-Cols D
      • Jaurrieta N
      • et al.
      Personality dimensions in obsessive–compulsive disorder: Relation to clinical variables.
      ,
      • Lyoo IK
      • Lee DW
      • Kim YS
      • Kong SW
      • Kwon JS
      Patterns of temperament and character in subjects with obsessive-compulsive disorder.
      ). Additionally, we observed depression-like behaviors in AD mice, a common characteristic present both in AD and OCD conditions (
      • Abramowitz JS
      • Storch EA
      • Keeley M
      • Cordell E
      Obsessive-compulsive disorder with comorbid major depression: What is the role of cognitive factors?.
      ,
      • Tibi L
      • van Oppen P
      • van Balkom AJLM
      • Eikelenboom M
      • Rickelt J
      • Schruers KRJ
      • Anholt GE
      The long-term association of OCD and depression and its moderators: A four-year follow up study in a large clinical sample.
      ,
      • Filali M
      • Lalonde R
      • Rivest S
      Cognitive and non-cognitive behaviors in an APPswe/PS1 bigenic model of Alzheimer’s disease.
      ,
      • Ledo JH
      • Azevedo EP
      • Clarke JR
      • Ribeiro FC
      • Figueiredo CP
      • Foguel D
      • et al.
      Amyloid-β oligomers link depressive-like behavior and cognitive deficits in mice [no. 10].
      ). Although anxiety is comorbid with OCD patients, our current AD model shows intact anxiety behaviors. Similarly, AD mice show no difference in locomotion and entries into the center in the open field experiment. These observations exclude the possibility of the impact of anxiety and hyperactivity on the compulsive behaviors exhibited by Aβ1-42 induced AD mice. Taken together, our behavioral results suggest that AD and OCD status share some overlapping cognitive symptoms that are consistent with recent clinical observations (
      • Dondu A
      • Sevincoka L
      • Akyol A
      • Tataroglu C
      Is Obsessive–Compulsive symptomatology a risk factor for Alzheimer-type dementia?.
      ,
      • Mrabet Khiari H
      • Achouri A
      • Ben Ali N
      • Cherif A
      • Batti H
      • Messaoud T
      • Mrabet A
      Obsessive-compulsive disorder: a new risk factor for Alzheimer disease?.
      ).
      The cortico-striatal circuit is critically involved in mediating compulsive behavior in humans (
      • Graybiel AM
      • Rauch SL
      Toward a Neurobiology of Obsessive-Compulsive Disorder.
      ,
      • Aouizerate B
      • Guehl D
      • Cuny E
      • Rougier A
      • Bioulac B
      • Tignol J
      • Burbaud P
      Pathophysiology of obsessive–compulsive disorder.
      ) and rodents (
      • Graybiel AM
      • Rauch SL
      Toward a Neurobiology of Obsessive-Compulsive Disorder.
      ,
      • Milad MR
      • Rauch SL
      Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
      ,
      • Menzies L
      • Chamberlain SR
      • Laird AR
      • Thelen SM
      • Sahakian BJ
      • Bullmore ET
      Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
      ,
      • Ahmari SE
      • Dougherty DD
      Dissecting Ocd Circuits: From Animal Models to Targeted Treatments.
      ,
      • Fettes P
      • Schulze L
      • Downar J
      Cortico-Striatal-Thalamic Loop Circuits of the Orbitofrontal Cortex: Promising Therapeutic Targets in Psychiatric Illness.
      ). Recent findings show that alterations in the functional connectivity of neuronal networks is associated with amyloid deposition (
      • Ben-Nejma IRH
      • Keliris AJ
      • Daans J
      • Ponsaerts P
      • Verhoye M
      • Van der Linden A
      • Keliris GA
      Increased soluble amyloid-beta causes early aberrant brain network hypersynchronisation in a mature-onset mouse model of amyloidosis.
      ,
      • Ishibashi K
      • Ishiwata K
      • Toyohara J
      • Murayama S
      • Ishii K
      Regional analysis of striatal and cortical amyloid deposition in patients with Alzheimer’s disease.
      ). For instance, functional connectivity deficits in cortico-striatal brain networks have been demonstrated to be driven by soluble Aβ neurotoxicity in a mature transgenic mice model of amyloidosis (
      • Paterson D
      • Nordberg A
      Neuronal nicotinic receptors in the human brain.
      ). Furthermore, in APP/PS1 mice that exhibit compulsive-like behavior as early as 3-5 months of age, structural and functional alterations to cortico-striatal synchronization have been reported (
      • Peng Y
      • Cai P
      • Hu J
      • Jiang J
      • Zhang J
      • Liu K
      • et al.
      Altered corticostriatal synchronization associated with compulsive-like behavior in APP/PS1 mice.
      ). There are evidences to show the role of other brain regions, for instance, the thalamus and amygdala (
      • Ullrich M
      • Weber M
      • Post AM
      • Popp S
      • Grein J
      • Zechner M
      • et al.
      OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency.
      ) in the pathology of OCD. A study using SPRED2 knockout mice revealed that dysfunction of the thalamo-amygdala circuit also contributes to the pathogenesis of OCD (
      • Ullrich M
      • Weber M
      • Post AM
      • Popp S
      • Grein J
      • Zechner M
      • et al.
      OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency.
      ). Interestingly, recent studies have shown the involvement of the HPC (
      • Milad MR
      • Rauch SL
      Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
      ,
      • Menzies L
      • Chamberlain SR
      • Laird AR
      • Thelen SM
      • Sahakian BJ
      • Bullmore ET
      Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
      ,
      • Reess TJ
      • Rus OG
      • Gürsel DA
      • Schmitz-Koep B
      • Wagner G
      • Berberich G
      • Koch K
      Association between hippocampus volume and symptom profiles in obsessive–compulsive disorder.
      ,
      • Wood J
      • Ahmari SE
      A Framework for Understanding the Emerging Role of Corticolimbic-Ventral Striatal Networks in OCD-Associated Repetitive Behaviors.
      ), in the pathophysiology of OCD. A functional connection exists between the HPC and mPFC and their communication is reflected in oscillatory coherence (
      • Colgin LL
      Oscillations and hippocampal–prefrontal synchrony.
      ,
      • Sigurdsson T
      • Duvarci S
      Hippocampal-Prefrontal Interactions in Cognition, Behavior and Psychiatric Disease.
      ). It has been suggested that disturbances in the oscillatory interaction between HPC and mPFC contribute to cognitive deficits in neuropsychiatric and neurodegenerative disorders (
      • Colgin LL
      Oscillations and hippocampal–prefrontal synchrony.
      ). However, the link between the alterations of oscillatory interactions within the hippocampo-cortical circuit and compulsive behavior seen in AD still remain unclear. In the present study, we noticed a considerable decrease in the power of theta and gamma band frequencies in the HPC and mPFC of AD mice under awake condition. Both the regions exhibited a statistically significant difference in oscillation power, with the exception of hippocampal gamma oscillation. Notably, we observed impairment in the coherence at the theta frequency in the HPC-mPFC neuronal circuit. A reorganization of hippocampal and cortical networks is well documented in AD patients and animal models. This reorganization is triggered by an imbalance between excitation and inhibition, resulting in impaired network activity (
      • Goutagny R
      • Krantic S
      Hippocampal Oscillatory Activity in Alzheimer’s Disease: Toward the Identification of Early Biomarkers?.
      ). Moreover, hippocampus and prefrontal cortex function is reported to be reduced in people at clinical high risk (CHR) of psychosis (
      • Allen P
      • Seal ML
      • Valli I
      • Fusar-Poli P
      • Perlini C
      • Day F
      • et al.
      Altered prefrontal and hippocampal function during verbal encoding and recognition in people with prodromal symptoms of psychosis.
      ,
      • Modinos G
      • Kempton MJ
      • Tognin S
      • Calem M
      • Porffy L
      • Antoniades M
      • et al.
      Association of Adverse Outcomes With Emotion Processing and Its Neural Substrate in Individuals at Clinical High Risk for Psychosis.
      ). Taken together, the disrupted LFP oscillations found in the HPC and mPFC and the impaired theta coherence between these regions in AD mice may indicate dysfunction of the hippocampo-cortical circuit.
      Accumulating evidence suggests that impairment of neurotransmitter systems could contribute to the dysfunction of neuronal networks and lead to an imbalance of inhibition/excitation, which profoundly affects cognition. AD-associated dementia is known to be associated with the dysregulation of cholinergic (
      • Hampel H
      • Mesulam M-M
      • Cuello AC
      • Farlow MR
      • Giacobini E
      • Grossberg GT
      • et al.
      The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease.
      ) and glutamatergic systems (
      • Liu J
      • Chang L
      • Song Y
      • Li H
      • Wu Y
      The Role of NMDA Receptors in Alzheimer’s Disease.
      ). The cholinergic system of the brain consists mainly of nicotinic acetylcholine receptors (
      • Kalamida D
      • Poulas K
      • Avramopoulou V
      • Fostieri E
      • Lagoumintzis G
      • Lazaridis K
      • et al.
      Muscle and neuronal nicotinic acetylcholine receptors: Structure, function and pathogenicity.
      ) and muscarinic acetylcholine receptors (
      • Scarr E
      Muscarinic Receptors: Their Roles in Disorders of the Central Nervous System and Potential as Therapeutic Targets.
      ), which are members of the cys-loop superfamily of ligand-gated ion channels (
      • Connolly CN
      • Wafford KA
      The Cys-loop superfamily of ligand-gated ion channels: the impact of receptor structure on function.
      ) and G protein-coupled receptors (
      • Wess J
      • Eglen RM
      • Gautam D
      Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development [no. 9].
      ), respectively. These receptors have also been identified in the brain areas implicated in OCD (
      • Hampel H
      • Mesulam M-M
      • Cuello AC
      • Farlow MR
      • Giacobini E
      • Grossberg GT
      • et al.
      The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease.
      ). There is evidence that the glutamatergic (
      • Arnold PaulD
      • Rosenberg DavidR
      • Mundo E
      • Tharmalingam S
      • Kennedy JamesL
      • MargaretA Richter
      Association of a glutamate (NMDA) subunit receptor gene (GRIN2B) with obsessive-compulsive disorder: a preliminary study.
      ,
      • Wu K
      • Hanna GL
      • Rosenberg DR
      • Arnold PD
      The role of glutamate signaling in the pathogenesis and treatment of obsessive–compulsive disorder.
      ) and cholinergic systems are implicated in the pathophysiology of OCD (
      • Lucey JV
      • Butcher G
      • Clare AW
      • Dinan TG
      Elevated growth hormone responses to pyridostigmine in obsessive-compulsive disorder: Evidence of cholinergic supersensitivity.
      ,
      • Smith EA
      • Russell A
      • Lorch E
      • Banerjee SP
      • Rose M
      • Ivey J
      • et al.
      Increased medial thalamic choline found in pediatric patients with obsessive-compulsive disorder versus major depression or healthy control subjects: a magnetic resonance spectroscopy study.
      ). Additionally, nicotine augmentation has been shown to improve clinical symptoms of OCD in recent studies, where patients with treatment-resistant OCD exhibited partial or no response to multiple selective serotonin reuptake inhibitors (SSRIs) (
      • Bergman J
      • Miodownik C
      • Lerner PP
      • Miodownik E
      • Shulkin A
      • Lerner V
      Donepezil as Add-on Treatment for Resistant Obsessive-Compulsive Disorder: Retrospective Case Series.
      ,
      • Pasquini M
      • Garavini A
      • Biondi M
      Nicotine augmentation for refractory obsessive-compulsive disorder. A case report.
      ). The gene GRIN2B that codes for the NR2B subunit of glutamate receptors in specific, is highly expressed within the striatum and PFC, and has been shown to be associated with predisposition to OCD (
      • Arnold PaulD
      • Rosenberg DavidR
      • Mundo E
      • Tharmalingam S
      • Kennedy JamesL
      • MargaretA Richter
      Association of a glutamate (NMDA) subunit receptor gene (GRIN2B) with obsessive-compulsive disorder: a preliminary study.
      ,
      • Arnold PD
      • MacMaster FP
      • Richter MA
      • Hanna GL
      • Sicard T
      • Burroughs E
      • et al.
      Glutamate receptor gene (GRIN2B) associated with reduced anterior cingulate glutamatergic concentration in pediatric obsessive-compulsive disorder.
      ). Furthermore, the majority of nAChR subtypes expressed in the brain areas linked to OCD are the nicotinic α4β2 receptors, encoded by CHRNA4 (
      • Mitra S
      • Mucha M
      • Khatri SN
      • Glenon R
      • Schulte MK
      • Bult-Ito A
      Attenuation of Compulsive-Like Behavior Through Positive Allosteric Modulation of α4β2 Nicotinic Acetylcholine Receptors in Non-Induced Compulsive-Like Mice.
      ,
      • Tsutsui-Kimura I
      • Ohmura Y
      • Izumi T
      • Yamaguchi T
      • Yoshida T
      • Yoshioka M
      Endogenous acetylcholine modulates impulsive action via α4β2 nicotinic acetylcholine receptors in rats.
      ). The CHRNA7 (codes for α7 nAChR) that is highly expressed within the hippocampus, is known to functionally interact with Aβ42 (
      • Wang HY
      • Lee DH
      • D’Andrea MR
      • Peterson PA
      • Shank RP
      • Reitz AB
      beta-Amyloid(1-42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer’s disease pathology.
      ), and is being increasingly considered as a target gene for schizophrenia and psychosis in AD (
      • Carson R
      • Craig D
      • Hart D
      • Todd S
      • McGuinness B
      • Johnston JA
      • et al.
      Genetic variation in the alpha 7 nicotinic acetylcholine receptor is associated with delusional symptoms in Alzheimer’s disease.
      ). In light of this, the present study targeted assessing the possible changes in the expression patterns of muscarinic acetylcholine receptors (M1, M3, M5), nicotinic acetylcholine receptors (α4, α7), and glutamate (NR2B) receptors in the dorsal and ventral hippocampus as well as mPFC of AD mice. We also assessed the expression of PLCβ1, an important enzyme involved in the acetylcholine signaling pathway. Interestingly, regulation of theta oscillations by the cholinergic-PLC signaling pathway plays a major role in hippocampal functions and diverse behaviors (
      • Shin J
      • Gireesh G
      • Kim S-W
      • Kim D-S
      • Lee S
      • Kim Y-S
      • et al.
      Phospholipase C 4 in the Medial Septum Controls Cholinergic Theta Oscillations and Anxiety Behaviors.
      ,
      • Gu Z
      • Alexander GM
      • Dudek SM
      • Yakel JL
      Hippocampus and Entorhinal Cortex Recruit Cholinergic and NMDA Receptors Separately to Generate Hippocampal Theta Oscillations.
      ,
      • Shin J
      • Kim D
      • Bianchi R
      • Wong RKS
      • Shin H-S
      Genetic dissection of theta rhythm heterogeneity in mice.
      ). Recent evidences suggest that hippocampal interneuronal α7 nAChRs regulate theta oscillations in freely moving mice (
      • Gu Z
      • Smith KG
      • Alexander GM
      • Guerreiro I
      • Dudek SM
      • Gutkin B
      • et al.
      Hippocampal Interneuronal α7 nAChRs Modulate Theta Oscillations in Freely Moving Mice.
      ). Our molecular experiments clearly revealed a downregulation in the expression patterns of both cholinergic and glutamate transmission receptors in the HPC and mPFC of AD mice compared to the control mice. These results explain the impairment in the coherence at theta frequency in the HPC-mPFC neuronal circuit in AD mice. Altogether, these results indicate that therapies involving muscarinic and nicotinic acetylcholine receptors could be an alternative approach for attenuating compulsive-like behaviors associated with AD, thereby posing important translational potential. Future studies are expected to establish neural mechanisms underlying how the interaction of different neurotransmitters affect compulsive behaviours in the AD state. For instance, the implication of the ​brain serotonin system​ in​ OCD has been a​ leading​ hypothesis in the ​recent decades​, ​primarily attributable to the relatively high effectiveness of the treatment of SSRIs (
      • Kim M
      • Kwak S
      • Yoon YB
      • Kwak YB
      • Kim T
      • Cho KIK
      • et al.
      Functional connectivity of the raphe nucleus as a predictor of the response to selective serotonin reuptake inhibitors in obsessive-compulsive disorder [no. 12].
      ). Although the studies are limited, there is convincing evidence of abnormalities in central norepinephrine (NE) function in patients with compulsive disorders (
      • Lustberg D
      • Iannitelli AF
      • Tillage RP
      • Pruitt M
      • Liles LC
      • Weinshenker D
      Central norepinephrine transmission is required for stress-induced repetitive behavior in two rodent models of obsessive-compulsive disorder.
      ).
      Interestingly, growing evidence suggests that neuro-inflammation could drive the pathogenic process in AD. The analysis of Alzheimer's patients with mild cognitive impairment further supports early and significant involvement of inflammation in disease pathogenesis (
      • Heneka MT
      • Carson MJ
      • Khoury JE
      • Landreth GE
      • Brosseron F
      • Feinstein DL
      • et al.
      Neuroinflammation in Alzheimer’s disease.
      ). Specifically in AD, it has been shown that there is an elevated expression of inflammatory mediators in the vicinity of Aβ peptide deposits and neurofibrillary tangles, suggesting the relationship between neuroinflammation and neurodegeneration (
      • Kinney JW
      • Bemiller SM
      • Murtishaw AS
      • Leisgang AM
      • Salazar AM
      • Lamb BT
      Inflammation as a central mechanism in Alzheimer’s disease.
      ). Additional studies are required to define if there is a relationship between neuroinflammation in AD and compulsive behaviors.
      In summary, our findings demonstrate perturbations to the physiological and molecular interactions between HPC and mPFC in the Aβ1-42 induced mouse model of AD. It further provides a strong impetus for exploring the potential role of other brain regions outside the cortico-striatal circuit in AD-associated compulsive-like behaviour. Future studies using behavioral state-dependent and targeted circuit manipulations are required to assess how the hippocampo-medial prefrontal cortex neuronal circuit dysfunction can play a role in compulsive-like behaviors during AD condition.

      ACKNOWLEDGMENTS AND DISCLOSURE

      This work was supported by Science and Engineering Research Board (SERB), India (No. CRG/2020/004205) and Ramalingaswami Re-entry Fellowship, Department of Biotechnology, India (No. BT/RLF/Re-863 entry/49/2018) to GG. ABS was supported by the Innovation in Science Pursuit for Inspired Research (INSPIRE) fellowship, Department of Science and Technology, India (IF190783). SFM was supported by Dr. TMA Pai Ph.D. scholarship, Manipal Academy of Higher Education (MAHE), Manipal, India. The authors thank Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), and Manipal for the infrastructure and support. The authors report no biomedical financial interests or potential conflicts of interest.

      Supplementary Material

      REFERENCES

        • Geda YE
        • Roberts RO
        • Knopman DS
        • Petersen RC
        • Christianson TJH
        • Pankratz VS
        • et al.
        Prevalence of Neuropsychiatric Symptoms in Mild Cognitive Impairment and Normal Cognitive Aging: Population-Based Study.
        Arch Gen Psychiatry. 2008; 65: 1193
        • Lanctôt KL
        • Amatniek J
        • Ancoli-Israel S
        • Arnold SE
        • Ballard C
        • Cohen-Mansfield J
        • et al.
        Neuropsychiatric signs and symptoms of Alzheimer’s disease: New treatment paradigms.
        Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2017; 3: 440-449
        • Rosenberg PB
        • Nowrangi MA
        • Lyketsos CG
        Neuropsychiatric symptoms in Alzheimer’s disease: What might be associated brain circuits?.
        Molecular Aspects of Medicine. 2015; 43–44: 25-37
        • Dondu A
        • Sevincoka L
        • Akyol A
        • Tataroglu C
        Is Obsessive–Compulsive symptomatology a risk factor for Alzheimer-type dementia?.
        Psychiatry Research. 2015; 225: 381-386
        • Mrabet Khiari H
        • Achouri A
        • Ben Ali N
        • Cherif A
        • Batti H
        • Messaoud T
        • Mrabet A
        Obsessive-compulsive disorder: a new risk factor for Alzheimer disease?.
        Neurol Sci. 2011; 32: 959-962
        • Ruggeri M
        • Ricci M
        • Gerace C
        • Blundo C
        Late-onset obsessive-compulsive disorder as the initial manifestation of possible behavioural variant Alzheimer’s disease.
        Cognitive Neuropsychiatry. 2022; 27: 11-19
        • Robbins TW
        • Vaghi MM
        • Banca P
        Obsessive-Compulsive Disorder: Puzzles and Prospects.
        Neuron. 2019; 102: 27-47
        • Graybiel AM
        • Rauch SL
        Toward a Neurobiology of Obsessive-Compulsive Disorder.
        Neuron. 2000; 28: 343-347
        • Milad MR
        • Rauch SL
        Obsessive-compulsive disorder: beyond segregated cortico-striatal pathways.
        Trends in Cognitive Sciences. 2012; 16: 43-51
        • Shmelkov SV
        • Hormigo A
        • Jing D
        • Proenca CC
        • Bath KG
        • Milde T
        • et al.
        Slitrk5 deficiency impairs corticostriatal circuitry and leads to obsessive-compulsive–like behaviors in mice [no. 5].
        Nat Med. 2010; 16: 598-602
        • Welch JM
        • Lu J
        • Rodriguiz RM
        • Trotta NC
        • Peca J
        • Ding J-D
        • et al.
        Cortico-striatal synaptic defects and OCD-like behaviours in Sapap3-mutant mice.
        Nature. 2007; 448: 894-900
        • Boedhoe PSW
        • Schmaal L
        • Abe Y
        • Ameis SH
        • Arnold PD
        • Batistuzzo MC
        • et al.
        Distinct Subcortical Volume Alterations in Pediatric and Adult OCD: A Worldwide Meta- and Mega-Analysis.
        AJP. 2017; 174: 60-69
        • Menzies L
        • Chamberlain SR
        • Laird AR
        • Thelen SM
        • Sahakian BJ
        • Bullmore ET
        Integrating evidence from neuroimaging and neuropsychological studies of obsessive-compulsive disorder: The orbitofronto-striatal model revisited.
        Neurosci Biobehav Rev. 2008; 32: 525-549
        • Rao S
        • Raveendranathan D
        • Shivakumar V
        • Narayanaswamy JC
        • Venkatasubramanian G
        • Reddy YCJ
        Hippocampus volume alterations and the clinical correlates in medication naïve obsessive compulsive disorder.
        Journal of Affective Disorders. 2018; 236: 1-5
        • Reess TJ
        • Rus OG
        • Gürsel DA
        • Schmitz-Koep B
        • Wagner G
        • Berberich G
        • Koch K
        Association between hippocampus volume and symptom profiles in obsessive–compulsive disorder.
        NeuroImage: Clinical. 2018; 17: 474-480
        • Schmitz TW
        • Correia MM
        • Ferreira CS
        • Prescot AP
        • Anderson MC
        Hippocampal GABA enables inhibitory control over unwanted thoughts [no. 1].
        Nat Commun. 2017; 8: 1311
        • Ullrich M
        • Weber M
        • Post AM
        • Popp S
        • Grein J
        • Zechner M
        • et al.
        OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency.
        Mol Psychiatry. 2018; 23: 444-458
        • Deacon RMJ
        • Croucher A
        • Rawlins JNP
        Hippocampal cytotoxic lesion effects on species-typical behaviours in mice.
        Behavioural Brain Research. 2002; 132: 203-213
        • Deacon RMJ
        • Penny C
        • Rawlins JNP
        Effects of medial prefrontal cortex cytotoxic lesions in mice.
        Behavioural Brain Research. 2003; 139: 139-155
        • Colgin LL
        Oscillations and hippocampal–prefrontal synchrony.
        Current Opinion in Neurobiology. 2011; 21: 467-474
        • O’Neill P-K
        • Gordon JA
        • Sigurdsson T
        Theta Oscillations in the Medial Prefrontal Cortex Are Modulated by Spatial Working Memory and Synchronize with the Hippocampus through Its Ventral Subregion.
        Journal of Neuroscience. 2013; 33: 14211-14224
        • Ruggiero RN
        • Rossignoli MT
        • Marques DB
        • de Sousa BM
        • Romcy-Pereira RN
        • Lopes-Aguiar C
        • Leite JP
        Neuromodulation of Hippocampal-Prefrontal Cortical Synaptic Plasticity and Functional Connectivity: Implications for Neuropsychiatric Disorders.
        Front Cell Neurosci. 2021; 15732360
        • Sun T
        • Song Z
        • Tian Y
        • Tian W
        • Zhu C
        • Ji G
        • et al.
        Basolateral amygdala input to the medial prefrontal cortex controls obsessive-compulsive disorder-like checking behavior.
        Proc Natl Acad Sci U S A. 2019; 116: 3799-3804
      1. Stine WB, Jungbauer L, Yu C, LaDu MJ (2010): Preparing Synthetic Aβ in Different Aggregation States. In: Roberson ED, editor. Alzheimer’s Disease and Frontotemporal Dementia, vol. 670. Totowa, NJ: Humana Press, pp 13–32.

        • Wang L-S
        • Tao X
        • Liu X-M
        • Zhou Y-F
        • Zhang M-D
        • Liao Y-H
        • et al.
        Cajaninstilbene Acid Ameliorates Cognitive Impairment Induced by Intrahippocampal Injection of Amyloid-β1–42 Oligomers.
        Front Pharmacol. 2019; 10: 1084
        • Gangadharan G
        • Shin J
        • Kim S-W
        • Kim A
        • Paydar A
        • Kim D-S
        • et al.
        Medial septal GABAergic projection neurons promote object exploration behavior and type 2 theta rhythm.
        Proc Natl Acad Sci USA. 2016; 113: 6550-6555
        • Shin J
        • Gireesh G
        • Kim S-W
        • Kim D-S
        • Lee S
        • Kim Y-S
        • et al.
        Phospholipase C 4 in the Medial Septum Controls Cholinergic Theta Oscillations and Anxiety Behaviors.
        Journal of Neuroscience. 2009; 29: 15375-15385
        • Kusunoki K
        • Sato T
        • Taga C
        • Yoshida Y
        • Komori K
        • Narita T
        • et al.
        Low novelty-seeking differentiates obsessive-compulsive disorder from major depression: Low novelty-seeking in OCD.
        Acta Psychiatrica Scandinavica. 2000; 101: 403-405
        • Abreu-Villaça Y
        • Queiroz-Gomes F do E
        • Dal Monte AP
        • Filgueiras CC
        • Manhães AC
        Individual differences in novelty-seeking behavior but not in anxiety response to a new environment can predict nicotine consumption in adolescent C57BL/6 mice.
        Behavioural Brain Research. 2006; 167: 175-182
        • Abramowitz JS
        • Storch EA
        • Keeley M
        • Cordell E
        Obsessive-compulsive disorder with comorbid major depression: What is the role of cognitive factors?.
        Behaviour Research and Therapy. 2007; 45: 2257-2267
        • Tibi L
        • van Oppen P
        • van Balkom AJLM
        • Eikelenboom M
        • Rickelt J
        • Schruers KRJ
        • Anholt GE
        The long-term association of OCD and depression and its moderators: A four-year follow up study in a large clinical sample.
        Eur psychiatr. 2017; 44: 76-82
        • Pearson-Leary J
        • McNay EC
        Intrahippocampal Administration of Amyloid-β 1-42 Oligomers Acutely Impairs Spatial Working Memory, Insulin Signaling, and Hippocampal Metabolism.
        Journal of Alzheimer’s Disease. 2012; 30: 413-422
        • van der Wee NJA
        • Ramsey NF
        • Jansma JM
        • Denys DA
        • van Megen HJGM
        • Westenberg HMG
        • Kahn RS
        Spatial working memory deficits in obsessive compulsive disorder are associated with excessive engagement of the medial frontal cortex.
        NeuroImage. 2003; 20: 2271-2280
        • Mehak SF
        • Shivakumar AB
        • Kumari S
        • Muralidharan B
        • Gangadharan G
        Theta and gamma oscillatory dynamics in mouse models of Alzheimer’s disease: A path to prospective therapeutic intervention.
        Neurosci Biobehav Rev. 2022; 136104628
        • Lei H
        • Lai J
        • Sun X
        • Xu Q
        • Feng G
        Lateral orbitofrontal dysfunction in the Sapap3 knockout mouse model of obsessive–compulsive disorder.
        jpn. 2019; 44: 120-131
        • Pogarell O
        • Juckel G
        • Mavrogiorgou P
        • Mulert C
        • Folkerts M
        • Hauke W
        • et al.
        Symptom-specific EEG power correlations in patients with obsessive–compulsive disorder.
        International Journal of Psychophysiology. 2006; 62: 87-92
        • Ahmari SE
        • Dougherty DD
        Dissecting Ocd Circuits: From Animal Models to Targeted Treatments.
        Depression and Anxiety. 2015; 32: 550-562
        • Fettes P
        • Schulze L
        • Downar J
        Cortico-Striatal-Thalamic Loop Circuits of the Orbitofrontal Cortex: Promising Therapeutic Targets in Psychiatric Illness.
        Front Syst Neurosci. 2017; 11: 25
        • Wood J
        • Ahmari SE
        A Framework for Understanding the Emerging Role of Corticolimbic-Ventral Striatal Networks in OCD-Associated Repetitive Behaviors.
        Front Syst Neurosci. 2015; 9: 171
        • Cummings JL
        • Back C
        The Cholinergic Hypothesis of Neuropsychiatric Symptoms in Alzheimer’s Disease.
        The American Journal of Geriatric Psychiatry. 1998; 6: S64-S78
        • Lanari A
        • Amenta F
        • Silvestrelli G
        • Tomassoni D
        • Parnetti L
        Neurotransmitter deficits in behavioural and psychological symptoms of Alzheimer’s disease.
        Mechanisms of Ageing and Development. 2006; 127: 158-165
        • Lucey JV
        • Butcher G
        • Clare AW
        • Dinan TG
        Elevated growth hormone responses to pyridostigmine in obsessive-compulsive disorder: Evidence of cholinergic supersensitivity.
        The American Journal of Psychiatry. 1993; 150: 961-962
        • Mitra S
        • Mucha M
        • Khatri SN
        • Glenon R
        • Schulte MK
        • Bult-Ito A
        Attenuation of Compulsive-Like Behavior Through Positive Allosteric Modulation of α4β2 Nicotinic Acetylcholine Receptors in Non-Induced Compulsive-Like Mice.
        Frontiers in Behavioral Neuroscience. 2017; 10: 244
        • Piacentino D
        • Maraone A
        • Roselli V
        • Berardelli I
        • Biondi M
        • Kotzalidis GD
        • Pasquini M
        Efficacy of nicotine administration on obsessions and compulsions in OCD: a systematic review.
        Annals of General Psychiatry. 2020; 19: 57
        • Yeung JHY
        • Calvo-Flores Guzmán B
        • Palpagama TH
        • Ethiraj J
        • Zhai Y
        • Tate WP
        • et al.
        Amyloid-beta1–42 induced glutamatergic receptor and transporter expression changes in the mouse hippocampus.
        Journal of Neurochemistry. 2020; 155: 62-80
        • Tsutsui-Kimura I
        • Ohmura Y
        • Izumi T
        • Yamaguchi T
        • Yoshida T
        • Yoshioka M
        Endogenous acetylcholine modulates impulsive action via α4β2 nicotinic acetylcholine receptors in rats.
        European Journal of Pharmacology. 2010; 641: 148-153
        • Luigjes J
        • Segrave R
        • de Joode N
        • Figee M
        • Denys D
        Efficacy of Invasive and Non-Invasive Brain Modulation Interventions for Addiction.
        Neuropsychol Rev. 2019; 29: 116-138
        • Frydman I
        • Ferreira-Garcia R
        • Borges MC
        • Velakoulis D
        • Walterfang M
        • Fontenelle LF
        Dementia Developing in Late-onset and Treatment-refractory Obsessive-compulsive Disorder.
        Cognitive and Behavioral Neurology. 2010; 23: 205-208
        • Chamberlain SR
        • Fineberg NA
        • Blackwell AD
        • Clark L
        • Robbins TW
        • Sahakian BJ
        A neuropsychological comparison of obsessive–compulsive disorder and trichotillomania.
        Neuropsychologia. 2007; 45: 654-662
        • Purcell R
        • Maruff P
        • Kyrios M
        • Pantelis C
        Cognitive Deficits in Obsessive–Compulsive Disorder on Tests of Frontal–Striatal Function.
        Biological Psychiatry. 1998; 43: 348-357
        • Kasza Á
        • Penke B
        • Frank Z
        • Bozsó Z
        • Szegedi V
        • Hunya Á
        • et al.
        Studies for Improving a Rat Model of Alzheimer’s Disease: Icv Administration of Well-Characterized β-Amyloid 1-42 Oligomers Induce Dysfunction in Spatial Memory [no. 11].
        Molecules. 2017; 22: 2007
        • Zhu H
        • Yan H
        • Tang N
        • Li X
        • Pang P
        • Li H
        • et al.
        Impairments of spatial memory in an Alzheimer’s disease model via degeneration of hippocampal cholinergic synapses [no. 1].
        Nat Commun. 2017; 8: 1676
        • Filali M
        • Lalonde R
        • Rivest S
        Cognitive and non-cognitive behaviors in an APPswe/PS1 bigenic model of Alzheimer’s disease.
        Genes, Brain and Behavior. 2009; 8: 143-148
        • Gimenez-Llort L
        • Alveal-Mellado D
        Digging Signatures in 13-Month-Old 3xTg-AD Mice for Alzheimer’s Disease and Its Disruption by Isolation Despite Social Life Since They Were Born.
        Frontiers in Behavioral Neuroscience. 2021; 14611384
        • Peng Y
        • Cai P
        • Hu J
        • Jiang J
        • Zhang J
        • Liu K
        • et al.
        Altered corticostriatal synchronization associated with compulsive-like behavior in APP/PS1 mice.
        Experimental Neurology. 2021; 344113805
        • Shepherd A
        • May C
        • Churilov L
        • Adlard PA
        • Hannan AJ
        • Burrows EL
        Evaluation of attention in APP/PS1 mice shows impulsive and compulsive behaviours.
        Genes, Brain and Behavior. 2021; 20e12594
        • Torres-Lista V
        • López-Pousa S
        • Giménez-Llort L
        Marble-burying is enhanced in 3xTg-AD mice, can be reversed by risperidone and it is modulable by handling.
        Behavioural Processes. 2015; 116: 69-74
        • Torres-Lista V
        • Giménez-Llort L
        Impairment of nesting behaviour in 3xTg-AD mice.
        Behavioural Brain Research. 2013; 247: 153-157
        • Alonso P
        • Menchón JM
        • Jiménez S
        • Segalàs J
        • Mataix-Cols D
        • Jaurrieta N
        • et al.
        Personality dimensions in obsessive–compulsive disorder: Relation to clinical variables.
        Psychiatry Research. 2008; 157: 159-168
        • Lyoo IK
        • Lee DW
        • Kim YS
        • Kong SW
        • Kwon JS
        Patterns of temperament and character in subjects with obsessive-compulsive disorder.
        J Clin Psychiatry. 2001; 62: 637-641
        • Ledo JH
        • Azevedo EP
        • Clarke JR
        • Ribeiro FC
        • Figueiredo CP
        • Foguel D
        • et al.
        Amyloid-β oligomers link depressive-like behavior and cognitive deficits in mice [no. 10].
        Mol Psychiatry. 2013; 18: 1053-1054
        • Aouizerate B
        • Guehl D
        • Cuny E
        • Rougier A
        • Bioulac B
        • Tignol J
        • Burbaud P
        Pathophysiology of obsessive–compulsive disorder.
        Progress in Neurobiology. 2004; 72: 195-221
        • Ben-Nejma IRH
        • Keliris AJ
        • Daans J
        • Ponsaerts P
        • Verhoye M
        • Van der Linden A
        • Keliris GA
        Increased soluble amyloid-beta causes early aberrant brain network hypersynchronisation in a mature-onset mouse model of amyloidosis.
        Acta Neuropathologica Communications. 2019; 7: 180
        • Ishibashi K
        • Ishiwata K
        • Toyohara J
        • Murayama S
        • Ishii K
        Regional analysis of striatal and cortical amyloid deposition in patients with Alzheimer’s disease.
        European Journal of Neuroscience. 2014; 40: 2701-2706
        • Paterson D
        • Nordberg A
        Neuronal nicotinic receptors in the human brain.
        Progress in Neurobiology. 2000; 61: 75-111
        • Sigurdsson T
        • Duvarci S
        Hippocampal-Prefrontal Interactions in Cognition, Behavior and Psychiatric Disease.
        Frontiers in Systems Neuroscience. 2016; 9: 190
        • Goutagny R
        • Krantic S
        Hippocampal Oscillatory Activity in Alzheimer’s Disease: Toward the Identification of Early Biomarkers?.
        Aging Dis. 2013; 4: 134-140
        • Allen P
        • Seal ML
        • Valli I
        • Fusar-Poli P
        • Perlini C
        • Day F
        • et al.
        Altered prefrontal and hippocampal function during verbal encoding and recognition in people with prodromal symptoms of psychosis.
        Schizophr Bull. 2011; 37: 746-756
        • Modinos G
        • Kempton MJ
        • Tognin S
        • Calem M
        • Porffy L
        • Antoniades M
        • et al.
        Association of Adverse Outcomes With Emotion Processing and Its Neural Substrate in Individuals at Clinical High Risk for Psychosis.
        JAMA Psychiatry. 2020; 77: 190-200
        • Hampel H
        • Mesulam M-M
        • Cuello AC
        • Farlow MR
        • Giacobini E
        • Grossberg GT
        • et al.
        The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease.
        Brain. 2018; 141 (–1933): 1917
        • Liu J
        • Chang L
        • Song Y
        • Li H
        • Wu Y
        The Role of NMDA Receptors in Alzheimer’s Disease.
        Frontiers in Neuroscience. 2019; 13: 43
        • Kalamida D
        • Poulas K
        • Avramopoulou V
        • Fostieri E
        • Lagoumintzis G
        • Lazaridis K
        • et al.
        Muscle and neuronal nicotinic acetylcholine receptors: Structure, function and pathogenicity.
        FEBS Journal. 2007; 274: 3799-3845
        • Scarr E
        Muscarinic Receptors: Their Roles in Disorders of the Central Nervous System and Potential as Therapeutic Targets.
        CNS Neuroscience & Therapeutics. 2012; 18: 369-379
        • Connolly CN
        • Wafford KA
        The Cys-loop superfamily of ligand-gated ion channels: the impact of receptor structure on function.
        Biochemical Society Transactions. 2004; 32: 529-534
        • Wess J
        • Eglen RM
        • Gautam D
        Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development [no. 9].
        Nat Rev Drug Discov. 2007; 6: 721-733
        • Arnold PaulD
        • Rosenberg DavidR
        • Mundo E
        • Tharmalingam S
        • Kennedy JamesL
        • MargaretA Richter
        Association of a glutamate (NMDA) subunit receptor gene (GRIN2B) with obsessive-compulsive disorder: a preliminary study.
        Psychopharmacology. 2004; 174: 530-538
        • Wu K
        • Hanna GL
        • Rosenberg DR
        • Arnold PD
        The role of glutamate signaling in the pathogenesis and treatment of obsessive–compulsive disorder.
        Pharmacology Biochemistry and Behavior. 2012; 100: 726-735
        • Smith EA
        • Russell A
        • Lorch E
        • Banerjee SP
        • Rose M
        • Ivey J
        • et al.
        Increased medial thalamic choline found in pediatric patients with obsessive-compulsive disorder versus major depression or healthy control subjects: a magnetic resonance spectroscopy study.
        Biological Psychiatry. 2003; 54: 1399-1405
        • Bergman J
        • Miodownik C
        • Lerner PP
        • Miodownik E
        • Shulkin A
        • Lerner V
        Donepezil as Add-on Treatment for Resistant Obsessive-Compulsive Disorder: Retrospective Case Series.
        Clin Neuropharm. 2016; 39: 194-196
        • Pasquini M
        • Garavini A
        • Biondi M
        Nicotine augmentation for refractory obsessive-compulsive disorder. A case report.
        Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2005; 29: 157-159
        • Arnold PD
        • MacMaster FP
        • Richter MA
        • Hanna GL
        • Sicard T
        • Burroughs E
        • et al.
        Glutamate receptor gene (GRIN2B) associated with reduced anterior cingulate glutamatergic concentration in pediatric obsessive-compulsive disorder.
        Psychiatry Res. 2009; 172: 136-139
        • Wang HY
        • Lee DH
        • D’Andrea MR
        • Peterson PA
        • Shank RP
        • Reitz AB
        beta-Amyloid(1-42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer’s disease pathology.
        J Biol Chem. 2000; 275: 5626-5632
        • Carson R
        • Craig D
        • Hart D
        • Todd S
        • McGuinness B
        • Johnston JA
        • et al.
        Genetic variation in the alpha 7 nicotinic acetylcholine receptor is associated with delusional symptoms in Alzheimer’s disease.
        Neuromolecular Med. 2008; 10: 377-384
        • Gu Z
        • Alexander GM
        • Dudek SM
        • Yakel JL
        Hippocampus and Entorhinal Cortex Recruit Cholinergic and NMDA Receptors Separately to Generate Hippocampal Theta Oscillations.
        Cell Reports. 2017; 21: 3585-3595
        • Shin J
        • Kim D
        • Bianchi R
        • Wong RKS
        • Shin H-S
        Genetic dissection of theta rhythm heterogeneity in mice.
        Proc Natl Acad Sci USA. 2005; 102: 18165-18170
        • Gu Z
        • Smith KG
        • Alexander GM
        • Guerreiro I
        • Dudek SM
        • Gutkin B
        • et al.
        Hippocampal Interneuronal α7 nAChRs Modulate Theta Oscillations in Freely Moving Mice.
        Cell Reports. 2020; 31107740
        • Kim M
        • Kwak S
        • Yoon YB
        • Kwak YB
        • Kim T
        • Cho KIK
        • et al.
        Functional connectivity of the raphe nucleus as a predictor of the response to selective serotonin reuptake inhibitors in obsessive-compulsive disorder [no. 12].
        Neuropsychopharmacol. 2019; 44 (–2081): 2073
        • Lustberg D
        • Iannitelli AF
        • Tillage RP
        • Pruitt M
        • Liles LC
        • Weinshenker D
        Central norepinephrine transmission is required for stress-induced repetitive behavior in two rodent models of obsessive-compulsive disorder.
        Psychopharmacology. 2020; 237 (–1987): 1973
        • Heneka MT
        • Carson MJ
        • Khoury JE
        • Landreth GE
        • Brosseron F
        • Feinstein DL
        • et al.
        Neuroinflammation in Alzheimer’s disease.
        The Lancet Neurology. 2015; 14: 388-405
        • Kinney JW
        • Bemiller SM
        • Murtishaw AS
        • Leisgang AM
        • Salazar AM
        • Lamb BT
        Inflammation as a central mechanism in Alzheimer’s disease.
        Alzheimer’s & Dementia: Translational Research & Clinical Interventions. 2018; 4: 575-590