Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 Choline supplementation in early life improves and low levels of choline can impair outcomes in a mouse model of Alzheimer’s disease. Elissavet Chartampila1,2, Karim S. Elayouby1,3, Paige Leary1,4, John J. LaFrancois1,5, David Alcantara-Gonzalez1,5, Swati Jain1, Kasey Gerencer1,6, Justin J. Botterill1,7, Stephen D. Ginsberg1,4,8,9, and Helen E. Scharfman1,4,5,8,9 1Center for Dementia Research The Nathan Kline Institute for Psychiatric Research Orangeburg, NY 10962 2 Current address: Department of Cell Biology and Physiology University of North Carolina Chapel Hill, NC 27510 3Current address: Department of Neurology Mount Sinai School of Medicine New York, NY 10029 4Department of Neuroscience and Physiology New York University Grossman School of Medicine New York, NY 100016 5Departments of Child and Adolescent Psychiatry New York University Grossman School of Medicine New York, NY 10016 6Current address: Department of Psychology University of Maine Orono, ME 04469 7Current address: Department of Anatomy, Physiology, & Pharmacology College of Medicine Saskatoon, SK S7N 5E5 8Department of Psychiatry New York University Grossman School of Medicine New York, NY 10016 9NYU Neuroscience Institute New York University Grossman School of Medicine New York, NY 10016 bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 Address for correspondence: Helen E. Scharfman Center for Dementia Research The Nathan Kline Institute for Psychiatric Research Orangeburg, NY 10962 Key words: dementia, maternal choline supplementation, hippocampus, dentate gyrus, hilus, interictal spike, novel object location, novel object recognition Word count: Abstract: 250 Introduction: 726 Discussion: 1705 Number of figures: 7 Number of supplemental figures: 4 Acknowledgements: Supported by NIH R01 AG055328 (HES, SDG), MH109305 (HES) and AG077103 (SDG) and the New York State Office of Mental Health (HES, JJL). We thank Dr. Christos Lisgaras for comments on the manuscript and Dr. Korey Kam for contributions at an early stage of this project. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 ABSTRACT Maternal choline supplementation (MCS) improves cognition in Alzheimer’s disease (AD) models. However, effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice, and many other mouse models and AD patients, are generalized EEG spikes (interictal spikes; IIS). Hyperexcitability is also reflected by elevated expression of the transcription factor ∆FosB in the principal cells of the dentate gyrus (DG), granule cells (GCs). We also studied the hilus of the DG because hilar neurons regulate GC excitability. We found reduced expression of the neuronal marker NeuN within hilar neurons in Tg2576 mice, which other studies have shown is a sign of oxidative stress or other pathology. Tg2576 breeding pairs received a diet with a relatively low, intermediate or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ∆FosB expression was reduced, and NeuN expression restored. Spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have negative consequences. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, and spatial memory in an animal model of AD. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 I. INTRODUCTION Diet has been suggested to influence several aspects of brain health. One dietary intervention that has been studied extensively in rodents and humans is supplementation of the maternal diet with the nutrient choline, (maternal choline supplementation; MCS). MCS improves many aspects of brain health in humans (Zeisel & da Costa 2009, Jiang et al. 2014). In normal rats, MCS also is beneficial, with numerous studies showing improved behavior in the adult offspring (for review, see (for review see Meck & Williams 2003)). In Alzheimer’s disease (AD), changes to the diet have been recommended (Bourre 2006, Power et al. 2019, Mao et al. 2021, Lobo et al. 2022), including MCS (Strupp et al. 2016, Velazquez et al. 2020). The recommendation to use MCS is based primarily on data from the Ts65Dn mouse model of Down syndrome (DS) and AD, where MCS led to improved memory and attention in Ts65Dn offspring (Strupp et al. 2016, Powers et al. 2017, Powers et al. 2021). In addition, degeneration of basal forebrain cholinergic neurons (BFCNs) in Ts65Dn mice, a hallmark of DS and AD, was reduced (Kelley et al. 2016, Powers et al. 2017). Based on the studies of Ts65Dn mice, we asked if MCS would improve another impairment that has been found in AD patients and mouse models of AD, hyperexcitability. We chose to study Tg2576 mice because hyperexcitability is robust, and BFCNs are likely to play a role (Kam et al. 2016, Lisgaras & Scharfman 2023). In our past work, the primary type of hyperexcitability was interictal spikes (IIS), which are named because they occur in between seizures (ictal events) in epilepsy. They are studied by EEG. We also studied hyperexcitability using ΔFosB expression, a transcription factor that is increased when neurons have been highly active over the prior 10-14 days (McClung et al. 2004). We studied ΔFosB in granule cells (GCs) of the dentate gyrus (DG), because numerous ΔFosB-expressing GCs occur when there are IIS (You et al. 2017, You et al. 2018). We also asked if behavior would improve if MCS reduced IIS frequency in Tg2576 mice. The basis for this question is in studies of epilepsy, where IIS disrupt cognition (Rausch et al. 1978, Aarts et al. 1984, Holmes & Lenck-Santini 2006, Kleen et al. 2010, Kleen et al. 2013, Gelinas et al. 2016) . Also, IIS in the Tg2576 mouse start early in life (by 1 month of age), before memory impairment, which occurs at 3-4 months of age (Duffy et al. 2015). Therefore, IIS might disrupt activity sufficiently to promote behavioral impairments. We previously found that the novel object location (NOL) task is impaired at 3-4 months of age in Tg2576 mice (Duffy et al. 2015), so we evaluated NOL, and a related task, novel object recognition (NOR; Vogel-Ciernia & Wood 2014). We also studied expression of NeuN, a neuronal marker that is reduced in numerous disease conditions (Buckingham et al. 2008, Kadriu et al. 2009, Matsuda et al. 2009, Duffy et al. 2011, Duffy et al. 2015). Here we report loss of NeuN-ir in the DG hilus of Tg2576 mice, which is important because impaired hilar neurons could cause GC hyperexcitability (Sperk et al. 2007, Scharfman & Myers 2012). We fed dams one of three diets, which were relatively low, intermediate or high in choline. The high choline diet provided levels of choline similar to other bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 studies of MCS in rodents (Meck et al. 1988, Loy et al. 1991, Holler et al. 1996, Meck & Williams 1999, Sandstrom et al. 2002, Mellott et al. 2004, Glenn et al. 2012, Kelley et al. 2019). After weaning the intermediate diet was used. Offspring were implanted with electrodes for EEG at 1 month of age, and 24 hr-long recordings began 1/month starting at 1 week after surgery. NOL and NOR were tested at 3 months and again at 6 months, and then we perfusion-fixed mice, sectioned the brains, and evaluated ΔFosB and NeuN protein expression (Figure 1A). The results showed a remarkable effect of the high choline diet. IIS and spatial memory were improved, as was ΔFosB and NeuN expression. Interestingly, the relatively low choline diet had mixed effects, reducing IIS frequency, but making memory, ΔFosB, and NeuN worse. The mice also died prematurely. In summary, we make a strong argument for choline supplementation in early life to improve outcomes in an AD model, and potentially AD. II. METHODS A. Animals All experimental procedures followed the guidelines set by the National Institute of Health (NIH) and were approved by the Institutional Animal Care and Use Committee (IACUC) of the Nathan Kline Institute for Psychiatric Research. Mice were housed in standard mouse cages with corn cob bedding and a 12-hr light-dark cycle. Food and water were provided ad libitum. Tg2576 mice express a mutant form of human APP (isoform 695) with a mutation found in a Swedish cohort (Lys670Arg, Met671Leu). The expression is driven by the hamster prion protein promoter. Mice were bred in-house from heterozygous Tg2576 males and non-transgenic female mice (C57BL6/SJL F1 hybrid). Wildtype (WT) mice were obtained from Jackson Laboratories (Stock# 100012). At weaning, genotypes were determined using a protocol to detect APP695. Breeding pairs were randomly assigned to receive one of three diets with different concentrations of choline chloride: 1.1 g/kg (AIN-76A, Dyets), 2.0 g/kg (Purina 5008, W.F. Fisher and Son Inc.) and 5.0 g/kg (AIN-76A, Dyets; Supplemental Table 1). These diets (relatively “low choline” diet, “intermediate choline” diet and “high choline” diet, respectively) were used until weaning (25-30 days of age). After weaning, all mice were fed the intermediate diet. Mice were housed with others of the same sex (1-4 per cage). B. Behavior 1. General information. Mice were housed in the room where they would be tested behaviorally starting at least 24 hr prior to all experiments. Both NOL and NOR included three acclimations (5 min each) followed by a training and a testing session (5 min each; Figure 1B-D). The interval between training and testing was 60 minutes and therefore tested short-term memory (Vogel-Ciernia & Wood 2014). NOL and bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 NOR tests were separated by 7 days to ensure effects of one task did not affect the next. Prior studies have failed to find an effect on one task on the next when a 7 day-long interval is used (Botterill et al. 2021). The order of testing (NOL before NOR or NOR before NOL) was randomized. Video recordings of all training and testing sessions were captured using a USB camera (Logitech HD Pro C920, Logitech). Acclimations were conducted between 10:00 a.m. and 12:00 p.m., while training and testing was conducted between 1:00 p.m. and 4:00 p.m. All equipment was cleaned using 70% ethanol before each use. In pilot studies, we chose objects that would be explored equally by animals. Indeed, animals showed no preference for either of the objects (i.e., LEGO object and pineapple, Supplemental Figure 1) during training, as reported in the Results. 2. NOL NOL was conducted in a standard rat cage (26 cm wide x 40 cm long x 20 cm high) with different pictures on 3 sides of the cage to provide a consistent context and therefore foster acclimation. Pictures included several shapes and colors. The dimensions of the pictures were 1) 10 cm x 21 cm, 2) 16 cm x 18 cm, and 3) 17 cm x 20 cm. During the acclimations, animals were allowed to freely explore the cage. During the training session, mice were placed in the cage where they had been acclimated, with two identical objects, 1 in a corner of the cage (e.g., left top) and the second in the adjacent corner (e.g., left bottom, Supplemental Figure 1). The mice were then removed and placed in their home cage for 1 hr. During the test session, one of the objects was moved to the opposite end of the cage (left top to right top; Supplemental Figure 1). The two objects were identical pineapple-like objects made of metal that were painted and approximately 3 cm x 3 cm x 5 cm (Supplemental Figure 1; Botterill et al. 2021). 3. NOR NOR was conducted in a standard rat cage as described above. During the acclimations, animals were allowed to freely explore the cage. During the training session, mice were placed in the cage used for acclimation with two identical objects centered along the shortest cage wall (Supplemental Figure 1). The mouse was then removed and placed in their home cage for 1 hr. During the testing session, one of the objects was replaced with a different one. The objects that were identical were 2 pineapple-like objects, and the new object was made of 2 green LEGO pieces approximately 5 cm x 7 cm x 7 cm (Supplemental Fig. 1; Botterill et al. 2021). 4. Quantification The experimenter who conducted the analysis was blind to the genotype of the offspring and which object was novel or familiar. Videos of the training and testing sessions were analyzed offline. Exploration was quantified based on the guidelines of Vogel-Ciernia & Wood (2014), but in addition to time spent exploring, the number of approaches to an object was also quantified. Time spent exploring each object was defined as the duration of time the nose was bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 pointed at the object and the nose was located within 2 cm of the object. In addition, exploration time included the time animals spent on top of the object if they were looking down and sniffing it. An approach was defined by a movement toward the object and ending at least 2 cm from the edge of the object. Animals that remembered the objects explored during the training phase were expected to demonstrate an increased preference for novelty in the test phase (Ennaceur & Delacour 1988). In other words, exploration of the novel object during testing was expected to be higher than 50% of the total time of object exploration. C. Anatomy 1. Perfusion-fixation and sectioning. Mice were initially anesthetized by isoflurane inhalation (NDC# 07-893-1389, Patterson Veterinary), followed by urethane (2.5 g/kg; i.p.; Cat# U2500, SigmaAldrich). Under deep anesthesia, the abdominal and heart cavities were opened and the animal was transcardially-perfused using 10 mL of room temperature (RT) saline (0.9% sodium chloride in double distilled (dd) H2O) using a peristaltic pump (Minipuls1; Gilson), followed by 30 mL of 4oC 4% paraformaldehyde (PFA, Cat# 19210, Electron Microscopy Sciences) in 0.1 M phosphate buffer (PB; pH 7.4). The brains were immediately extracted, hemisected, and post-fixed for at least 24 hr in 4% PFA (at 4oC). Following post-fixation, the left hemisphere was sectioned in the coronal plane and the right hemisphere was sectioned in the horizontal plane (50 µmthick sections) using a vibratome (VT 1000P, Leica Biosystems). Sections were collected serially and stored at 4oC in 24-well tissue culture plates containing cryoprotectant solution (30% glycerol, 30% ethylene glycol, 40% 0.1 M phosphate buffer, pH 6.7). Throughout the hippocampus, every 6th section was processed with an antibody to NeuN or ΔFosB. 2. NeuN and ∆FosB immunohistochemistry Free floating sections were washed in 0.1 M Tris Buffer (TB; 3 x 5 min), followed by a 3 min wash in 1% H2O2. Sections were then washed in 0.1 M TB (3 x 5 min) and incubated for 60 min in 5% horse serum for NeuN (Cat# S-2000, Vector) or 5% goat serum for ∆FosB (Cat# S-1000, Vector), diluted in a solution of 0.25% (volume/volume or v/v) Triton-X 100, and 1% (weight/volume or w/v) bovine serum albumin in 0.1 M TB. Sections were then incubated overnight at 4oC in primary antibody to NeuN (mouse monoclonal, 1:5000; Cat# MAB377, Chemicon International) or anti-ΔFosB (rabbit monoclonal, 1:1000; Cat# D3S8R, Cell Signaling), diluted in a solution of 0.25% (v/v) Triton-X 100, and 1% (w/v) bovine serum albumin in 0.1M TB. Both NeuN and ΔFosB have been wellcharacterized as antigens (Mullen et al. 1992, Wolf et al. 1996, Chen et al. 1997, Sarnat et al. 1998) and the antibodies we used have been commonly employed in the past (Chen et al. 1997, Duffy et al. 2013, Corbett et al. 2017). On the following day, sections were washed in 0.1 M TB (3 x 5 min) and then incubated for 60 min in biotinylated horse anti-mouse secondary antibody for NeuN (1:500, Cat# BA-2000, Vector) or biotinylated goat anti-rabbit bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 secondary antibody for ∆FosB (1:500, Cat# BA-1000), diluted in a solution of 0.25% (v/v) Triton-X 100, and 1% (w/v) bovine serum albumin in 0.1 M TB. The sections were then washed in 0.1 M TB (3 x 5 min) and incubated in avidin-biotin complex for 2 hrs (1:1000; Cat# PK-6100, Vector). They were washed in 0.1 M TB (3 x 5 min) and then reacted in a solution containing 0.5 mg/mL 3, 3’diaminobenzidine (DAB; Cat# D5905, Sigma-Aldrich), 40 µg/mL ammonium chloride (Cat# A4514, Sigma-Aldrich), 25 mg/mL D(+)-glucose (Cat# G5767, Sigma-Aldrich), and 3 g/mL glucose oxidase (Cat# G2133, Sigma-Aldrich) in 0.1 M TB. This method slowed the reaction time so that the reaction could be temporally controlled, meaning that the reaction could be stopped when the immunoreactivity was robust but background was still low. The sections were then washed in 0.1 M TB (3 x 5 min), mounted on gelatin-coated slides (1% bovine gelatin, Cat# G9391, Sigma-Aldrich) and dried at RT overnight. The following day they were dehydrated in increasing concentrations of ethanol (90%, 10 min; 95% 10 min; 100%, 10 min; 100% again, 10 min), washed in Xylene (10 min; Cat# 534056, Sigma-Aldrich) and cover-slipped with Permount (Cat# 1798601, Electron Microscopy Sciences). 2. Analysis Photomicrographs were acquired using ImagePro Plus V7.0 (Media Cybernetics) and a digital camera (Model RET 2000R-F-CLR-12, Q-Imaging). NeuN and ∆FosB staining were quantified from micrographs using ImageJ (V1.44, National Institutes of Health). All images were first converted to grayscale and in each section, the hilus was traced, defined by zone 4 of Amaral (1978). A threshold was then calculated to identify the NeuN-stained cell bodies but not background. Then NeuN-stained cell bodies in the hilus were quantified manually. Note that the threshold was defined in ImageJ using the distribution of intensities in the micrograph. A threshold was then set to identify the dark cells but not the background slide using A slider was pushed from the low level of staining corresponding to background to the location where staining intensity makes a sharp rise, reflecting strongly stained cells. Below that was considered background and above that was defined as above threshold. To count ∆FosB-stained cells, images were converted to grayscale and in each section, the dentate gyrus GCL was outlined and defined as a region of interest (ROI). A threshold was then set to identify the dark cells but not the background, analogous to the method to define threshold in the preceding paragraph. Two thresholds were used. The first threshold was most inclusive of immunoreactive cells. Presumably this group reflected cells with less neuronal activity. Then counts were made a second time with a higher threshold. This group probably corresponded to cells with the highest activity. ImageJ was used to calculate the area (in pixels) within the ROI that was above threshold. D. Video-electroencephalographic (video-EEG) recordings 1. Stereotaxic surgery EEG electrodes (stainless steel jeweler’s screws) were implanted in Tg2576 animals at 4 weeks of age. WT littermates were not implanted because previous bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 work from our laboratory has shown that WT mice have no IIS (Kam et al. 2016). The animals were anesthetized by isoflurane inhalation (3% isoflurane. 2% oxygen for induction). For maintenance during surgery, isoflurane was < 1.75% and flow rate was 1.2 L/min. Mice were placed in a stereotaxic apparatus (David Kopf Instruments). Prior to the implantation of the electrodes, animals were injected with the analgesic Buprenex (0.2 mg/kg, s.c.; buprenorphine hydroxide, NDC# 12496-0757-5, Reckitt Benckiser), which was diluted in saline (0.03 mg/mL in sterile 0.9% sodium chloride solution, Vedco Inc.). The skull was exposed with a midline incision and six holes were drilled for the placement of subdural screw electrodes (Cat# 8209, 0.10” stainless steel screws, Pinnacle Technology). The coordinates for the electrode placement were: right occipital cortex (AP 3.5 mm, ML 2.0 mm), left frontal cortex (AP -0.5 mm, ML -1.5 mm), left hippocampus (AP -2.5 mm, ML -2.0 mm), and right hippocampus (AP -2.5 mm, ML 2.0 mm (Kam et al. 2016). Two additional screws were placed, one over the right olfactory bulb as ground (AP 2.3 mm, ML 1.8 mm) and another at the midline over the cerebellum as a reference (relative to Lambda: AP -1.5 mm, ML -0.5 mm). Screws were attached to an eight-pin connector (Cat# ED85100-ND, Digi-Key Corporation), which was placed over the skull and secured with dental cement (Cat# 4734FIB, Lang Dental Mfg. Co., Inc.). After surgery, animals were placed on a heating blanket overnight and injected with lactated Ringer’s solution (50 mL/kg at 31oC; NDC# 099355000476, Aspen Veterinary Resources Ltd). They were then transferred to the room where the video-EEG is recorded. Implanted animals were allowed to recover for one week following the surgery. 2. Video-EEG recording Video-EEG was recorded stating at 1.25 months (5 weeks) of age. The animals were then recorded at 2, 3, 4, 5 and 6 months of age. Each recording session lasted 24 hr so that a long period of sleep could be acquired, since IIS occur primarily in sleep (Kam et al. 2016). Mice were placed into 21 cm x 19 cm transparent cages with food and water provided ad libitum and corncob bedding. A pre-amplifier was inserted into the eight-pin connector on the skull, which was connected to a multichannel commutator (Cat# 8408, Pinnacle Technology). This arrangement allowed for free range of movement throughout the recording. EEG signals were acquired at 2000 Hz and bandpass filtered at 0.5 – 200 Hz using Sirenia Acquisition )V2.0.4, Pinnacle Technology). Simultaneous video recordings were captured using an infrared camera (Cat# AP-DCS100W, Apex CCTV). 3. Analysis EEG recordings were analyzed offline with Neuroscore V3.2.1 (Data Science International). IIS were defined as large amplitude, 10-75 msec deflections occurring synchronously in all four leads (Kam et al. 2016). They were quantified by first setting an amplitude threshold (calculated separately for each recording) and a duration criterion (10-75 msec). To determine the amplitude threshold, we bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 calculated the root mean square (RMS) amplitude for noise in each recording during a 60 sec segment of baseline activity that did not include any artifacts. The threshold for IIS was set at 9 standard deviations above the RMS amplitude. This threshold was selected because it identified IIS extremely well and excluded movement or other artifacts. The IIS in one of the hippocampal channels was used because of data suggesting IIS begin in the hippocampus (Kam et al. 2016). Following the automatic analysis of the IIS, the software’s detection accuracy was verified manually for each recording to ensure that all spikes appeared in all four channels and that no artifacts were included in the final IIS count. E. Statistical comparisons Data are expressed as mean ± standard error of the mean (SEM). P-value was set to <0.05 prior to all experiments. Parametric data comparing two groups used unpaired two-tailed t-tests. For >2 groups, one-way ANOVA was used followed by Tukey-Kramer post-hoc tests that corrected for multiple comparisons. For data with two main factors, two-way ANOVA was followed by Tukey-Kramer post-hoc tests. Interactions are not reported unless they were significant. For the analysis of IIS frequency, a repeated measures ANOVA was not possible because animal numbers were not the same at all ages. This was due to mortality in low choline-treated mice or other unidentified reasons. Therefore, a mixed effects analysis was conducted. For non-parametric data, Mann-Whitney U tests were used to compare two groups and Kruskal-Wallis for >2 groups. The post-hoc tests were Dunn’s test for multiple comparisons. Tests were conducted using Prism (V 9.4.1, Graphpad Software). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 III. RESULTS A. Behavior NOL and the NOR, unlike other tasks previously used to evaluate cognitive deficits in >9 months-old Tg2576 mouse model (i.e., Morris Water Maze, Radial Arm Water Maze, Y-Maze; Yassine et al. 2013, Wolf et al. 2016), provided an opportunity to capture cognitive deficits in animals as young as three months-old (Duffy et al. 2015). Figure 1B-D shows a schematic of the specific experimental procedures for NOL and NOR. 1. NOL The results of the NOL task are presented in Figure 2. Exploration of the novel object is expressed as a percentage of the total exploration of both objects. During the training session, animals did not show preference for one object over the other and this result was independent of genotype. Thus, a two-way ANOVA revealed no main effects of genotype on novel object exploration during training at three (F(1,97) = 0.40, p = 0.53) or six months of age (F(1,70) = 1.94, p = 0.17). There also was no effect of maternal diet on training at three (F(2,97) = 0.06, p = 0.94) or six months of age (F(2,70) = 2.94, p = 0.06). Next, we asked whether Tg2576 mice were deficient in the test phase of NOL. First we studied the 3 months-old mice. We used a two-way ANOVA with genotype and task phase (training versus testing) as main factors. In animals that received the intermediate diet, a two-way ANOVA revealed a significant main effect of the phase of the task (training or testing; F(1,24) = 7.48, p = 0.01). Consistent with previous studies (Duffy et al. 2015), post-hoc analyses revealed a significant increase in the exploration of the novel object during testing in WT (p = 0.02) but not Tg2576 mice (p = 0.50; Figure 2A2). Thus, Tg2576 mice were impaired in NOL when fed the intermediate diet. In animals that received the high choline diet, a two-way ANOVA revealed a main effect of the task phase (F(1,36) = 48.23, p < 0.001). Post-hoc analyses revealed a significant increase in the exploration of the novel object during testing in both WT (p < 0.001) and Tg2576 mice (p < 0.001; Figure 2A3). Therefore, high choline supplementation improved memory in Tg2576 mice. In animals treated with the low choline diet, there was no effect of phase of the task (F(1,37) = 1.39, p = 0.25). These data suggesting that relatively low levels of choline during early life impaired spatial memory in both WT and Tg2576 mice (Figure 2A). Thus, low choline had a significant adverse effect. Adverse effects are further supported by survival plots showing that there was more mortality at earlier ages in offspring exposed to the low choline diet (Supplemental Figure 1). Figure 2B shows the results in 6 months-old WT and Tg2576 mice. In animals that received the intermediate diet, a two-way ANOVA showed no main effect of genotype (F(1,18) = 0.01, p = 0.92) or task phase (F(1,18) = 3.40, p = 0.08), revealing age-related memory deficits even in WT mice at six months of age (Figure 2B). In contrast, the high choline group showed a main effect of task phase (F(1,29) = 28.32, p < 0.001). Post-hoc analyses showed a significant bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 increase in the exploration of the novel object during testing in both WT (p < 0.001) and Tg2576 (p = 0.02; Figure 2B). In animals that received the relatively low choline diet, there was no effect of task phase (F(1,25) = 3.00, p = 0.10; Figure 2B). Thus, low choline had adverse effects on NOL in both WT and Tg2576 mice. Taken together, these results demonstrate the lasting beneficial effects of the high choline diet and adverse effects of low choline on offspring in a spatial memory task in Tg2576 as well as WT littermates. 2. NOR We first confirmed that animals did not show preference for one object over the other in training. A two-way ANOVA revealed no main effects of genotype at three months (F(1,71) = 2.59, p = 0.54) and no effect of diet (F(2,71) = 0.22, p = 0.81). The same result was obtained at six months of age (genotype: (F(1,66) = 0.11, p = 0.75); diet: (F(2,66) = 0.98, p = 0.38). Thus, novel object exploration during training approached 50% in all treatment groups, independent of genotype and maternal diet, and at both ages. Figure 3A shows the results of the testing phase of the NOR task in 3 months-old WT and Tg2576 mice. In animals that received the intermediate choline diet, a two-way ANOVA revealed a significant main effect of task phase (F(1,19) = 40.53, p < 0.01) and no effect of genotype (F(1,19) = 0.78, p = 0.39). Post-hoc analyses revealed a significant increase in the exploration of the novel object during testing compared to training for both WT (p < 0.01) and Tg2576 (p < 0.01; Figure 3A). These results suggest that Tg2576 did not have a deficit in NOR at 3 months of age. Mice fed the high choline diet also showed no deficit in NOR at 3 months of age. Thus, a two-way ANOVA revealed a main effect of task phase (F(1,26) = 17.51, p < 0.01) but not genotype (F(1,26) = 3.53, p = 0.07). Post-hoc analyses revealed a significant increase in the exploration of the novel object during testing in both WT (p < 0.01) and Tg2576 (p = 0.03; Figure 3A). In animals treated with the low choline diet in early life, there was no impairment in NOR at 3 months of age (task phase (F(1,26) = 22.61, p < 0.01; genotype (F(1,26) = 0.02, p = 0.89). There was a significant increase in the exploration of the novel object in testing both in WT (p = 0.03) and Tg2576 (p < 0.01). Figure 3B shows that there were no impairments at 6 months of age. In animals that received the intermediate diet, a two-way ANOVA revealed a significant main effect of task phase (F(1,19) = 13.65, p < 0.01) but no main effect of genotype (F(1,19) = 0.28, p = 0.60). Animals explored the novel object significantly more during testing in WT (p = 0.03) and Tg2576 mice (p = 0.04; Figure 3B). In animals that received the other diets, results were the same. For the high choline group, there was an effect of task phase (F(1,27) = 16.26, p < 0.01) but not genotype (F(1,27) = 0.24, p = 0.63) and a significant increase in the exploration of the novel object during testing in both WT (p = 0.04) and Tg2576 (p < 0.01; Figure 3B). Similarly, in animals treated with the low choline diet, there was a main effect of task phase (F(1,26) = 32.66, p < 0.01), no effect of genotype bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 (F(1, 6) = 0.05, p = 0.82), and more exploration of the novel object during the test phase (WT, p < 0.01; Tg2576, p < 0.01). Thus, at both 3 and 6 months of age, WT and Tg2576 mice performed well in the NOR task. These results suggest that young Tg2576 mice are less sensitive to NOR than NOL. The greater sensitivity to NOL is consistent with past demonstrations that the dentate gyrus contributes to NOL (Sahay et al. 2011, Kesner et al. 2015, Spyrka & Hess 2018, Vandrey et al. 2020, Gulmez Karaca et al. 2021, GoodSmith et al. 2022), and early signs of adverse effects in the dentate gyrus in mouse models of AD (impaired LTP; Jacobsen et al. 2006, altered gene expression; Palop et al. 2011). Our implementation of NOL may have increased the dentate gyrus-dependence of the task by making the object locations relatively close together, because the dentate gyrus is critical to distinguishing small differences in locations. In studies by Pofahl and colleagues, it was shown that distances between objects like those we used made NOL dentate gyrus-dependent (Pofahl et al. 2021). 3. Exploration time Total object exploration (TOE) was measured to address any effects of genotype or diet on the total duration of exploration of objects (Figure 4). a. 3 months of age For NOL at 3 months of age, there were effects of genotype (F(1,67) = 6.891, p = 0.01) but not maternal diet (F(2,87) = 0.67, p = 0.63; Figure 4A). TukeyKramer post-hoc tests showed that Tg2576 mice fed the low choline diet showed more exploration than WT mice fed the same diet but the effect was on the border of significance (p = 0.049; data not shown). Three months-old mice tested with NOR showed no significant effect of genotype (F(1,76) = 1.35, p = 0.25) or diet (F(2,76) = 0.30, p = 0.61; Figure 4C). b. 6 months of age 1) Exploration NOL and NOR showed significant effects of diet on exploration at 6 months of age (Figure 4B,D). For NOL, there was no effect of genotype (F(1,66) = 0.33; p = 0.57) but there was an effect of maternal diet (F(2,66) = 8.82; p = 0.0004). For NOR, the same was true (genotype: F(1,72) = 0.96, p = 0.33; diet: F(2,72) = 8.50, p = 0.0005). We then pooled genotypes to determine which diets differed. For NOL, mice treated with the relatively low choline diet had less exploration than mice fed the high choline diet (one-way ANOVA, F(2,77) = 6.90; p = 0.002; Tukey post-hoc test, p = 0.005; Figure 4B) and the mice that were fed the intermediate diet also had less exploration than mice fed the high choline diet (Tukey post-hoc test, p = 0.008; Figure 4B). Results for NOR were the same: mice treated with the low choline diet had less exploration than mice fed the high choline diet (one-way ANOVA, F(2,74) =4.81; p = 0.02; Tukey post-hoc test, p = 0.02; Figure 4D) and the mice that were fed the intermediate diet also had less exploration than mice fed the high choline diet (Tukey post-hoc test, p = 0.03; Figure 4D). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 2) Approaches To gain insight into the potential reason for differences in exploration, we measured total object approaches (TOA; Figure 4A-D). TOA was defined as the number of approaches to the familiar object + number of approaches to the novel object. There were no differences by genotype (two-way ANOVA, F(1,73) = 2.930, p = 0.09) or maternal diet (F(2,73) = 2.36, p = 0.10; Figure 4A-D). Taken together, the results indicate that, in 6 months-old mice, animals that received the high choline diet spent more time with objects at each approach. This may underlie increased object location memory in high choline-treated mice by increasing time for information processing during an approach. It may also help in the NOR task with object recognition memory, although it did not seem impaired in mice fed other diets. Another possibility is that as Tg2576 mice age they show compensatory changes that enhance memory. This has been suggested for procedural learning (Middei et al. 2004) but not for memory of objects. A third possibility is that the high choline diet reduces anxiety (Glenn et al. 2012, Langley et al. 2015, McCall et al. 2015). Reduced anxiety may lessen fear of exploring objects, and as a result, animals may spend more time with objects. For 3 months-old mice there were no effects of maternal diet on exploration, suggesting that increased time with objects for a given approach is a change that develops with age. To our knowledge, this age-related change has not been previously reported. It could be a strategy to cope with impairments that are developing as AD pathology manifests. B Anatomy 1. NeuN To further analyze the effects of dietary choline early in life, we used an antibody against a neuronal nuclear antigen, NeuN. Previous studies have found that reduced expression of NeuN often occurs in neurons after insults or injury (e.g., ischemia, toxicity and even aging; Lind et al. 2005, Portiansky et al. 2006, Buckingham et al. 2008, Kadriu et al. 2009, Matsuda et al. 2009, Won et al. 2009, Duffy et al. 2011). For example, when NeuN is reduced after a reduction in ankyrin-rich membrane spanning kinase D-interacting substrate of 220K, ARMS/Kidins, a protein critical to neurotrophin signaling, neurons in the entorhinal cortex demonstrate signs of toxicity such as pkynosis (Duffy et al. 2011, Duffy et al. 2015). Since NeuN is reduced when A levels are elevated (Wu et al. 2016) and NeuN is reduced in AD patients (Camporez et al. 2021), we used NeuN expression to ask if the high choline diet could rescue the loss of NeuN-ir. As shown in Figure 5, relatively weak NeuN-ir was observed in Tg2576 animals in the hilus, an area known to be highly vulnerable to insult and injury (Scharfman 1999). The most NeuN loss appeared to be in mice that received the low choline diet and the intermediate diet compared to the high choline diet (Figure 5B). Therefore, we conducted a two-way ANOVA with diet and septotemporal levels as main factors. To examine the septotemporal axis, bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 quantification from 2-3 dorsal coronal sections were averaged to provide a value for dorsal DG and 2-3 ventral horizontal sections were averaged for a ventral value. There was no effect of diet (F(2,30) = 2.11, p = 0.14) nor septotemporal level (F(1,30) = 0.02, p = 0.88) but there was a significant interaction of diet and septotemporal level (F(2,30) = 3.88, p = 0.04). A one-way ANOVA using dorsal values showed a significant effect of diet (F(2,21)= 7.58, p = 0.003) but this was not the case for ventral values (F(2,23) = 0.13; p = 0.88; Figure 5D). In the dorsal DG, there was less NeuN-ir in mice fed the low choline diet versus the high choline diet (Tukey-Kramer post hoc test, p = 0.03) and less NeuN in mice fed the intermediate vs high choline diet (p = 0.003; Figure 5D). The results suggest that choline enrichment protected hilar neurons from NeuN loss in Tg2576 mice. 2. ∆FosB To complement the information from the video-EEG recordings we used a marker of elevated neuronal activity, ∆FosB (Figure 6). ∆FosB is a truncated variant of the transcription factor FosB, which is increased by enhanced neuronal activity; ∆FosB has a half-life of approximately 8 days (Ulery-Reynolds et al., 2009), so when ∆FosB is elevated, it reflects increased neuronal activity over the last 10-14 days (McClung et al. 2004). Previous studies have shown that when the J20 mouse model of AD is examined with an antibody to ∆FosB, the GC layer shows extremely high levels of ∆FosB expression (Corbett et al. 2017), similar to a mouse with chronic spontaneous seizures (epilepsy; Morris et al. 2000). This is not surprising since the J20 mice have recurrent seizures (Palop et al. 2007). Therefore, we asked if Tg2576 mice would have robust ∆FosB in the GC layer, and choline supplementation would reduce it. There was strong expression of ∆FosB in Tg2576 GCs especially in mice fed the low choline diet (Figure 6). Two-way ANOVA showed a main effect of maternal diet (F(2,18) = 3.76, p = 0.04) but there was not a main effect of septotemporal level (F(1,18) = 1.3, p = 0.26). To clarify the effect of maternal diet, a one-way ANOVA was conducted. There was an effect of diet (F(2,9) = 10.74, p = 0.004) with post-hoc tests showing significantly less GC ΔFosB expression in high choline-treated mice compared to low choline (p = 0.003). Low choline-treated mice were not significantly different from mice that were fed the intermediate diet (p = 0.08). These data used dorsal sections so we repeated the analysis for the ventral DG, and there also was no significant effect of diet (F2,9) = 0.79, p = 0.52; Figure 6). ∆FosB quantification was repeated with a lower threshold to define ∆FosB-ir GCs (see Methods) and results were the same (Figure 6). One-way ANOVA showed a significant effect of diet (F(2,41) = 14.66, p < 0.0001), and low choline mice had greater ΔFosB-ir than high choline mice (p =0.003). Mice fed the intermediate diet had higher expression than mice fed the high choline diet (p = 0.0001). These data suggest that high choline in the diet early in life can reduce hyperexcitability of GCs in offspring later in life. In addition, low choline has an opposite effect, again suggesting this maternal diet has adverse effects. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 C. IIS Previous research has shown that Tg2576 mice exhibit IIS (Figure 7A) starting at very young ages: 4 weeks (Kam et al. 2016) or 6 weeks (Bezzina et al. 2015). Therefore, we performed video-EEG using cortical (left frontal cortex, right occipital cortex) and hippocampal (left and right) electrodes in WT and Tg2576 mice (Figure 7B-C). Animals were recorded for 24 hr each session so that there was sufficient time to capture major behavioral states (exploration, awake rest, and sleep). Consistent with previous studies (Bezzina et al. 2015, Kam et al. 2016) we observed IIS in Tg2576 mice but not WT littermates. As shown in Figure 7, animals that received the intermediate diet had a significantly higher number of IIS in the 24 hr recording periods compared to animals that received the high choline and the low choline diets. A mixed model analysis showed that there was a significant effect of age (F(2,37) = 3.38; p = 0.04) and maternal diet (F(2,36) = 8.12; p = 0.01). At the 1.25-month recording, Tukey post-hoc analyses showed that IIS frequency in animals treated with the intermediate diet was higher compared to animals treated with the low (p = 0.03) or high choline diets (p = 0.04). This also was true for 2 months and 3 months (low choline, high choline, p <0.05). At 4 months, the low choline groups had significantly reduced IIS frequency compared to the intermediate diet (p = 0.009) but not the high choline group (p = 0.98). At 5-6 months, IIS frequencies were not significantly different in the mice fed the maternal diets (all p > 0.05). As shown previously (Kam et al. 2016), and in Figure 7, at 4-6 months of age there was high variability in IIS frequency between animals. It is possible that IIS frequency becomes more variable because we found that seizures occur in the Tg2576 mice after 2 months of age (Kam et al. 2016), and there was a sharp increase in IIS during the day before and after a seizure (Supplemental Figure 3). D. Seizures and premature mortality in mice fed the low choline diet We found mice fed the low choline diet had greater ΔFosB-ir in GCs and the hilus showed very low NeuN-ir. Therefore we asked whether low cholinetreated mice had more seizures than mice fed the other diets. We recorded 8 mice by video-EEG for 5 days each (at 6 months of age) to examine seizures and found 2 mice from the low choline group had seizures (11 seizures over 2 of the 5 days in one mouse, 1 seizure in the other mouse) whereas none of the other mice had seizures (n=0/4, 2 intermediate and 2 high choline, data not shown). These values are probably an underestimate for the low choline group because many mice in this group appeared to die in a severe seizure prior to 6 months of age (Supplemental Figure 2). Therefore, the survivors at 6 months probably were the subset with few seizures. The reason that low choline-treated mice appeared to die in a seizure was that they were found in a specific posture in their cage which occurs when a severe seizure leads to death (Supplemental Figure 2). They were found in a prone posture with extended, rigid limbs (Supplemental Figure 2). Regardless of how the mice died, there was greater mortality in the low choline group compared to mice that had been fed the high choline diet (Supplemental Figure 2). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 E. Sex differences As shown in Figures 2-3, there appeared to be no sex differences in NOL or NOR. For confirmation, we studied NOL further, since that was the task that showed effects of diet. A three-way ANOVA with sex, genotype and task phase as factors showed no significant effect of sex at three months of age (low choline diet, F(1,35) = 0.0001; p = 0.99). Results were the same for the three months-old mice fed the intermediate diet (F(1,22) = 0.062; p = 0.81) and three months-old mice that were fed the high choline diet (F(1,34) = 0.43; p = 0.52). At six months of age there also were no significant effects of sex (low choline diet, F(1,23) = 0.34; p = 0.57; intermediate diet, F(1,16) = 2.49; p = 0.13; high choline diet, F(1,27) = 0.29; p = 0.87). Regarding IIS, we did not find sex differences (Supplemental Figure 4). A two-way ANOVA with sex and diet as factors showed no effect of sex (F(1,20) =2.48; p = 0.13) or diet (F(1,20) = 3.56; p = 0.07). Note that for this comparison we used the low choline and high choline groups only because sex information was only available for these mice. The results for months 2-3 also showed no effect of sex (2 months: F(1,17) = 0.64; p = 0.43; 3 months: F(1,15) = 0.19; p = 0.67). IV. DISCUSSION Summary This study showed that early life choline supplementation had several beneficial effects in Tg2576 mice. The high choline diet led to reduced IIS frequency, improved behavior in the NOL task, and improved expression of ΔFosB. We showed for the first time that Tg2576 mice have pathology in the hilus at six months of age in that hilar neurons are deficient in immunostaining with an anti-NeuN antibody. Tg2576 offspring on the high choline diet showed a restoration of hilar NeuN staining. Because hilar neurons play a role in NOL (Bui et al. 2018), it is possible that rescue of hilar neurons by the high choline diet led to improved spatial memory. Hilar neurons have been suggested to contribute to excitability of GCs (Sloviter 1994, Scharfman 1999, Scharfman & Myers 2012, Jinde et al. 2013, Bui et al. 2018, Botterill et al. 2019), so phenotypic rescue of hilar neurons may also have contributed to the ability of the high choline diet to reduce hyperexcitability. We also found surprising effects of treating mice with relatively low choline in the maternal diet. On the one hand, IIS frequency was reduced. However, these mice showed premature mortality. Furthermore, in WT mice where no impairments in NOL normally occur, WT mice fed the low choline diet were impaired. In Tg2576 mice, the group that had the low maternal choline diet had the highest ΔFosB expression and the lowest NeuN expression of any experimental group. Potential reasons for these surprising effects are discussed below. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 Benefits of high choline The results of this study are consistent with previous reports that prenatal or postnatal choline supplementation improves object memory. One study of iron deficiency showed that choline supplementation improved NOR (Kennedy et al. 2014). Wistar rats treated with a diet high in choline had improved NOR relative to rats that had been fed a diet low in choline (Moreno et al. 2013). In rats that were aged to 24 months, animals that had been fed a high choline diet showed improved NOR, but only in females (Glenn et al. 2008). Although few studies have examined the effects of MCS on hyperexcitability, our results showing the benefits of high choline are consistent with reports that a methyl-enriched diet (including choline but also betaine, folic acid, vitamin B12, L-methionine and zinc) reduced spike wave discharges in the offspring (Sarkisova et al. 2023) and audiogenic seizure severity (Poletaeva et al. 2014). Perinatal ethanol treatment that increased excitability was mitigated by choline chloride injection from postnatal day 10-30 (Grafe et al. 2022). To our knowledge our study is the first to show that MCS can exert effects on IIS and ΔFosB. However, prior studies have shown that MCS in rats improved memory after severe seizures. Using a convulsant to induce several hours of severe seizures (status epilepticus, SE), there is usually extensive hippocampal neuronal loss and when tested in the subsequent days and weeks, memory impairment. MCS reduced this impairment following SE in the Morris water maze (Yang et al. 2000, Holmes et al. 2002, Wong-Goodrich et al. 2011). Our results are consistent with previous studies of mouse models of AD. In the APP/PS1 mouse model, it was shown that life-long choline supplementation, to the level we used (5.0 g/kg) improved memory in aged mice compared to the 1.1 g/kg diet (Velazquez et al. 2019). Postnatal choline supplementation also improved memory in APP/PS1 mice (Wang et al. 2019). Other improvements were also shown in the APP/PS1 mice, such as improved cholinergic expression via upregulation of choline acetyltransferase (Mellott et al. 2017, Velazquez et al. 2019). Adverse effects of low choline The effects of the relatively low choline diet were surprising because several adverse effects were found. This is unexpected because 1.1 g/kg choline is not considered very low in several studies of the past, and some publications consider it normal (e.g., Lamoureux et al. 2008). The majority of past studies used rats (Meck et al. 1988, Meck & Williams 1999, 2003, Meck et al. 2007, Lamoureux et al. 2008), however, so there could be a species difference. Also, some of the past studies used the diet for a shorter timespan than we did (e.g., Lamoureux et al. 2008) because of evidence that perinatal choline supplementation exerts its maximal effects during critical periods (Meck & Williams 2003, Meck et al. 2007). It was also surprising to find high mortality in mice exposed to the low choline diet in early life. Also, the offspring of mothers fed the low choline diet had very high ΔFosB expression in the GCs, suggesting they had chronic seizures. The observation that mice died from seizures is consistent with the ΔFosB data. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 There also was weak NeuN in the hilus. When WT mice were tested, NOL was impaired in the low choline group. These data suggest that the offspring of mothers fed the low choline diet were unhealthy, possibly leading to or because of seizures. On the other hand, IIS were reduced in frequency in the same mice. There are several possible explanations for this apparent paradox. The most parsimonious explanation is the mice were unhealthy. If there was disruption of sleep, IIS might be reduced since IIS occur in sleep primarily. However, there is no evidence that MCS influences sleep in humans or laboratory animals. The one study that looked at the sleep/wake cycle in rats did not find effects (Cheng et al. 2008). A more radical explanation is that the intermediate diet promoted IIS rather than low choline reducing IIS. The idea here is that choline is not critical to IIS because both low and high choline groups had low IIS frequency compared to the intermediate group, at least for 3 months of life. Instead of choline, a constituent of the intermediate diet may have promoted IIS. The reason why the effect of diet waned over time could have been due to the fact that after weaning all animals had the intermediate diet. More seizures could explain premature mortality in the low choline group. However, why it would decrease IIS is not clear. One reason seizures could cause IIS to be reduced is that seizures in the Tg2576 mice typically started during sleep and woke mice from sleep. The arousal from sleep would interrupt the behavioral state when IIS usually occur as mentioned above. Also, seizures in the Tg2576 mice were followed by a depression of the EEG that would reduce IIS (postictal depression; Supplemental Figure 3). NeuN As mentioned above, NeuN is a neuronal nuclear antigen that can be phosphorylated, and when that occurs the antibody to NeuN no longer binds to NeuN. NeuN is phosphorylated in response to oxidative damage, brain injury, and toxicity (Lind et al. 2005). Therefore, it was of interest when we saw that hilar neurons of the dentate gyrus showed reduced NeuN in Tg2576 mice. Hilar neurons are mainly glutamatergic mossy cells and somatostatin (SOM)/neuropeptide Y (NPY)-expressing GABAergic neurons (HIPP cells; (Houser 2007, Scharfman & Myers 2012, Scharfman 2016), and both neuronal types are implicated in spatial memory functions of the GCs, as well as their excitability (Myers & Scharfman 2009, 2011, Scharfman & Myers 2012, Jinde et al. 2013, Scharfman 2016, Raza et al. 2017, Bui et al. 2018, GoodSmith et al. 2019, Li et al. 2021). If damaged by the high intracellular APP/A levels in young Tg2576 mice, the hilar neurons would be expected to show NeuN loss. This idea is consistent with deficits in SOM and NPY-stained cells (Chan-Palay 1987), indicating a vulnerability. The reason for rescue of dorsal hilar but not ventral hilar NeuN with the high choline diet is unclear. However, there was a trend for the high choline mice to have greater NeuN in ventral tissue sections. The greater sensitivity of dorsal neurons may be related to differences in dorsal and ventral dentate gyrus gene bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 expression patterns, since some of the genes that are differentially expressed could affect vulnerability to insult or injury (Cembrowski et al. 2016, Zhang et al. 2018). Resilience of vulnerable neuronal populations, especially in the hippocampus, is a topic of high interest in AD research. ∆FosB Given that seizures are associated with elevated GC expression of ∆FosB (Chen et al. 1997, McClung et al. 2004, Corbett et al. 2017, You et al. 2017), the differences in ∆FosB expression levels observed here could be the result of increased seizures in animals that received the low choline diet. Therefore, we suggest that the high ΔFosB in Tg2576 mice fed the low choline diet reflects more seizures than in the other two diets. ∆FosB is a transcription factor that is linked to cognition. In the J20 mouse model of AD, elevated ∆FosB in GCs led to reduced cognition, and when ∆FosB was selectively reduced the cognition improved (Corbett et al. 2017). Therefore, the reduction in ∆FosB by the high choline diet was important to show hyperexcitability was reduced and also important because it showed how high choline may benefit cognition. Additional considerations Although the low and high choline diets only had differences in choline, the intermediate diet had different amounts of choline as well as other constituents (Supplementary Table 1). Therefore, the only diet comparison with a difference restricted to choline is the low versus high choline diets (Supplemental Table 1). The intermediate diet was useful however, because numerous studies in AD mouse models employ this standard diet. Moreover, it may have promoted IIS as discussed above. Although we failed to detect sex differences, there may actually have been differences if females had been analyzed according to their stage of the estrous cycle at death. This possibility is raised by prior data showing that rats and mice during diestrus have low excitability in hippocampal slices; at other cycle stages hyperexcitability occurs (Scharfman et al. 2003). The Tg2576 mouse model is one of many murine models of AD, and as with all models there are inherent limitations. The Tg2576 model of cerebral amyloid overexpression lacks tau pathology and recapitulates familial AD, whereas the majority of AD is sporadic. However, MCS has now demonstrated structural/functional benefits in several AD-relevant models, namely Tg2576 (this report), APP-PS1 (Mellott et al. 2017), and Ts65Dn (Powers et al. 2016, Strupp et al. 2016, Powers et al. 2017, Alldred et al. 2021, Alldred et al. 2023). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 Figure 1. Schematic of the behavioral procedures for the NOL and NOR tasks. A. Experimental timeline. Dams were fed one of three diets for mating, gestation, and offspring consumed the diet until weaning at 25-30 days of age. At 1 monthold, offspring were implanted with electrodes for EEG and recorded for a 24 hr period at 1.2, 2, 3, 4, 5, and 6 months of age. At 3 and 6 months of age behavior was tested. At 6 months of age, after behavioral testing, mice were perfused, brains were sectioned, and sections were processed with antibodies against NeuN and ∆FosB. B. Prior to NOL or NOR, animals were acclimated to the testing arena. There were three acclimation sessions separated by 24hrs during which animals were allowed to freely explore for 5 min. C. In the NOL task, animals were placed in a cage with two identical objects and allowed to freely explore for 5 min (Training). After one hour they were brought back to the cage, where one object was displaced, and allowed to freely explore for 5 min (Testing). D. In the NOR task. animals were placed in a cage with two identical objects and allowed to freely explore for 5 min (Training). After one hour they were brought back to the cage, where one object was replaced, and allowed to freely explore for 5 min (Testing). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 875 876 877 878 879 880 881 882 883 884 885 886 887 888 Figure 2. Choline enrichment reduced spatial memory deficits. A. Three months-old mice. 1. WT and Tg2576 mice exposed to the low choline diet showed spatial memory deficits. 2. Tg2576 exposed to the intermediate diet showed spatial memory deficits but not WT. 3. Spatial memory was improved in animals exposed to the high choline diet. B. Six months-old mice. 1. WT and Tg2576 mice exposed to the low choline diet showed spatial memory deficits. 2. WT and Tg2576 mice exposed to the intermediate diet showed spatial memory deficits. 3. WT and Tg2576 mice exposed to the high choline diet had improved spatial memory. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 889 bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 890 891 892 893 894 895 896 897 898 Figure 3. WT and Tg2576 mice showed recognition memory regardless of diet. A. Three months-old mice. 1-3. WT and Tg2576 mice treated performed the NOR task. B. Six months-old mice. 1-3. WT and Tg2576 mice performed the NOR task. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 899 900 901 902 903 904 905 906 907 908 909 910 911 912 Figure 4. Tg2576 mice treated with the choline supplemented diet spent more time with the objects at each approach. A-B. NOL. There was no effect of diet on time exploring or approaches to the novel object in the NOL task at 3 months of age (A) but there were significant differences at 6 months of age (B). Mice exposed to the high choline diet spent more time exploring than mice that had been fed the low choline or the intermediate diet. C-D. NOR. There was no effect of diet on time exploring or approaches to the novel object in the NOR task at 3 months of age (C) but there were significant differences at 6 months of age (D). Mice that had been fed the high choline diet spent more time exploring than mice that had been fed the low choline or the intermediate diet. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 Figure 5. Choline supplementation improves NeuN immunoreactivity (ir) in hilar cells in Tg2576 animals A. A sagittal view of the brain illustrating the coronal sectioning plane. B. Representative images of NeuN-ir staining in the dorsal dentate gyrus of Tg2576 animals from each treatment group. C. Quantification methods. Representative images demonstrating the thresholding criteria established to quantify NeuN-ir. 1. A NeuN-stained section shows an inset (arrow) expanded at the top left to show 3 hilar cells. NeuN-ir cells are marked by blue arrows. NeuN-ir that was too close to background staining to be considered positive is marked by a green arrow. 2. After converting the image to grayscale, a threshold was set so that all cells stained well with NeuN antibody were red. The inset shows that the two cells that were darkest met the threshold while the weakly stained cell did not. D. Six months-old Tg2576 mice treated with the high choline diet had significantly more NeuN-ir cells in the hilus in the dorsal dentate gyrus (1) compared to other treatment groups but not ventral dentate gyrus (2). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 Figure 6. Choline supplementation reduced ∆FosB expression in dorsal GCs of Tg2576 mice. A. A sagittal view of the brain illustrating the coronal sectioning plane. B. Representative images of ∆FosB staining in the anterior dentate gyrus of Tg2576 animals from each treatment group. C. Quantification methods. Representative images demonstrating the thresholding criteria established to quantify ∆FosB. 1. A ∆FosB -stained section shows strongly-stained cells (white arrows). 2. A strict thresholding criteria was used to make only the darkest stained cells red. D. Six months-old Tg2576 mice treated with the choline supplemented diet had significantly more ∆FosB -ir pixels above threshold in the dorsal dentate gyrus (1) compared to other treatment groups but not ventral dentate gyrus (2). E. Methods are shown using a threshold that was less strict. 1. Some of the stained cells that were included are not as dark as those used for the strict threshold (white arrows). 2. All cells above the less conservative threshold are shown in red. F. With the less conservative threshold, the results were the same as D. Mice that were fed the high choline diet had less ΔFosB-ir pixels than the mice that were fed the other diets. However, this was true for dorsal (1) but not ventral (2) sections. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 956 957 958 959 960 961 962 963 964 965 966 967 968 969 Figure 7. The high choline diet reduced IIS frequency in Tg2576 animals. A. 1. Representative example of an IIS (red arrow). IIS were defined as occurring in all four channels (generalized) to distinguish them from focal spikes that can often be artifact. 2-3. The IIS shown in A1 is expanded. B. 1. Scatter plot of IIS frequency at each age of recording. 2. Means and sem are plotted. The high choline diet group had fewer IIS than the intermediate diet group at ages 1-3 months ($, p<0.05) and the low choline group had less IIS than the intermediate diet at ages 1-4 months (@, p<0.05). The high and low choline diet groups were significantly different at ages 3 and 4 months (*, p<0.05). bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 970 971 972 973 Supplemental Table 1. Comparison of the three diets used in this study. Choline Low choline 1.1g/kg Intermediate choline 2.0g/kg High choline 5.0g/kg Carbohydrate Protein Fat Thiamin Riboflavin Pyridoxine Niacin Folic Acid Biotin 690 177 120 0.020 0.600 0.700 3.000 0.200 0.020 570 236 170 0.015 0.005 0.006 0.780 0.003 0.002 690 177 120 0.020 0.600 0.700 3.000 0.200 0.020 bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 974 975 976 977 978 979 980 981 Supplemental Figure 1. Details of behavioral tasks. A standard mouse cage is diagrammed with the locations of objects inside it. For NOL, there were two pineapple-like objects and one was moved across the cage for the test session. For the NOR test session, one pineapple-like object was removed and an object made of LEGO was placed in the location where the original pineapple-like object had been. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 982 983 984 985 986 987 988 989 Supplemental Figure 2. Mortality was high in mice treated with the low choline diet. A. Survival curves are shown for mice fed the low choline diet and mice fed the high choline diet. B. A photo of a mouse after sudden unexplained death. The mouse was found in a posture consistent with death during a convulsive seizure. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 990 991 992 993 994 995 996 997 998 Supplemental Figure 3. IIS frequency before and after seizures. A. Representative EEG trace of a seizure in a 5 months-old Tg2576 mouse. B. IIS frequency was quantified from continuous video-EEG for 4 days. During the first 24 hrs, IIS frequency was low. The next day a seizure occurred (arrow), and IIS frequency increased approximately 5 fold, and for the next 24 hr period. On day 4, IIS frequency returned to lower levels. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 999 1000 1001 1002 1003 Supplemental Figure 4. IIS frequency was similar for each sex. A. At 1.2 months-old, female and male Tg2576 mice were not significantly different in IIS frequency for the low and high choline diets. B-C. There were no significant sex differences at 2 (B) and 3 (C) months of age. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 REFERENCES Aarts JH, Binnie CD, Smit AM & Wilkins AJ (1984) Selective cognitive impairment during focal and generalized epileptiform EEG activity. Brain. 107 ( Pt 1): 293-308. Alldred MJ, Lee SH & Ginsberg SD (2021) Adiponectin modulation by genotype and maternal choline supplementation in a mouse model of Down syndrome and Alzheimer's disease. J Clin Med. 10. Alldred MJ, Che S & Ginsberg SD (2023) Basal forebrain cholinergic neurons are vulnerable in a mouse model of Down syndrome and their molecular fingerprint is rescued by maternal choline supplementation. Faseb J. 37: e22944. Bezzina C, Verret L, Juan C, Remaud J, Halley H, Rampon C & Dahan L (2015) Early onset of hypersynchronous network activity and expression of a marker of chronic seizures in the Tg2576 mouse model of Alzheimer's disease. PLoS One. 10: e0119910. Botterill JJ, Lu YL, LaFrancois JJ, Bernstein HL, Alcantara-Gonzalez D, Jain S, Leary P & Scharfman HE (2019) An excitatory and epileptogenic effect of dentate gyrus mossy cells in a mouse model of epilepsy. Cell Rep. 29: 28752889 e2876. Botterill JJ, Vinod KY, Gerencer KJ, Teixeira CM, LaFrancois JJ & Scharfman HE (2021) Bidirectional regulation of cognitive and anxiety-like behaviors by dentate gyrus mossy cells in male and female mice. J Neurosci. 41: 24752495. Bourre JM (2006) Effects of nutrients (in food) on the structure and function of the nervous system: Update on dietary requirements for brain. Part 2 : Macronutrients. J Nutr Health Aging. 10: 386-399. Buckingham BP, Inman DM, Lambert W, Oglesby E, Calkins DJ, Steele MR, Vetter ML, Marsh-Armstrong N & Horner PJ (2008) Progressive ganglion cell degeneration precedes neuronal loss in a mouse model of glaucoma. J Neurosci. 28: 2735-2744. Bui AD, Nguyen TM, Limouse C, Kim HK, Szabo GG, Felong S, Maroso M & Soltesz I (2018) Dentate gyrus mossy cells control spontaneous convulsive seizures and spatial memory. Science. 359: 787-790. Camporez D, Belcavello L, Almeida JFF, Silva-Sena GG, Pimassoni LHS, Morelato RL, do Carmo Pimentel Batitucci M & de Paula F (2021) Positive association of a sirt1 variant and parameters of oxidative stress on Alzheimer's disease. Neurol Sci. 42: 1843-1851. Cembrowski MS, Wang L, Sugino K, Shields BC & Spruston N (2016) Hipposeq: A comprehensive RNA-seq database of gene expression in hippocampal principal neurons. Elife. 5: e14997. Chan-Palay V (1987) Somatostatin immunoreactive neurons in the human hippocampus and cortex shown by immunogold/silver intensification on vibratome sections: Coexistence with neuropeptide Y neurons, and effects in Alzheimer-type dementia. J Comp Neurol. 260: 201-223. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 Chen J, Kelz MB, Hope BT, Nakabeppu Y & Nestler EJ (1997) Chronic fosrelated antigens: Stable variants of ∆FosB induced in brain by chronic treatments. J Neurosci. 17: 4933-4941. Cheng RK, Williams CL & Meck WH (2008) Oscillatory bands, neuronal synchrony and hippocampal function: Implications of the effects of prenatal choline supplementation for sleep-dependent memory consolidation. Brain Res. 1237: 176-194. Corbett BF, You JC, Zhang X, Pyfer MS, Tosi U, Iascone DM, Petrof I, Hazra A, Fu CH, Stephens GS, Ashok AA, Aschmies S, Zhao L, Nestler EJ & Chin J (2017) ∆FosB regulates gene expression and cognitive dysfunction in a mouse model of Alzheimer's disease. Cell Rep. 20: 344-355. Duffy AM, Schaner MJ, Wu SH, Staniszewski A, Kumar A, Arevalo JC, Arancio O, Chao MV & Scharfman HE (2011) A selective role for ARMS/Kidins220 scaffold protein in spatial memory and trophic support of entorhinal and frontal cortical neurons. Exp Neurol. 229: 409-420. Duffy AM, Schaner MJ, Chin J & Scharfman HE (2013) Expression of c-fos in hilar mossy cells of the dentate gyrus in vivo. Hippocampus. 23: 649-655. Duffy AM, Morales-Corraliza J, Bermudez-Hernandez KM, Schaner MJ, Magagna-Poveda A, Mathews PM & Scharfman HE (2015) Entorhinal cortical defects in Tg2576 mice are present as early as 2-4 months of age. Neurobiol Aging. 36: 134-148. Ennaceur A & Delacour J (1988) A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behav Brain Res. 31: 47-59. Gelinas JN, Khodagholy D, Thesen T, Devinsky O & Buzsaki G (2016) Interictal epileptiform discharges induce hippocampal-cortical coupling in temporal lobe epilepsy. Nat Med. 22: 641-648. Glenn MJ, Kirby ED, Gibson EM, Wong-Goodrich SJ, Mellott TJ, Blusztajn JK & Williams CL (2008) Age-related declines in exploratory behavior and markers of hippocampal plasticity are attenuated by prenatal choline supplementation in rats. Brain Res. 1237: 110-123. Glenn MJ, Adams RS & McClurg L (2012) Supplemental dietary choline during development exerts antidepressant-like effects in adult female rats. Brain Res. 1443: 52-63. GoodSmith D, Lee H, Neunuebel JP, Song H & Knierim JJ (2019) Dentate gyrus mossy cells share a role in pattern separation with dentate granule cells and proximal CA3 pyramidal cells. J Neurosci. 39: 9570-9584. GoodSmith D, Kim SH, Puliyadi V, Ming GL, Song H, Knierim JJ & Christian KM (2022) Flexible encoding of objects and space in single cells of the dentate gyrus. Curr Biol. 32: 1088-1101 e1085. Grafe EL, Wade MMM, Hodson CE, Thomas JD & Christie BR (2022) Postnatal choline supplementation rescues deficits in synaptic plasticity following prenatal ethanol exposure. Nutrients. 14. Gulmez Karaca K, Brito DVC, Kupke J, Zeuch B & Oliveira AMM (2021) Engram reactivation during memory retrieval predicts long-term memory performance in aged mice. Neurobiol Aging. 101: 256-261. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 Hauser WA (1992) Seizure disorders: The changes with age. Epilepsia. 33 Suppl 4: S6-14. Holler T, Cermak JM & Blusztajn JK (1996) Dietary choline supplementation in pregnant rats increases hippocampal phospholipase d activity of the offspring. FASEB J. 10: 1653-1659. Holmes GL, Yang Y, Liu Z, Cermak JM, Sarkisian MR, Stafstrom CE, Neill JC & Blusztajn JK (2002) Seizure-induced memory impairment is reduced by choline supplementation before or after status epilepticus. Epilepsy Res. 48: 313. Holmes GL & Lenck-Santini PP (2006) Role of interictal epileptiform abnormalities in cognitive impairment. Epilepsy Behav. 8: 504-515. Houser CR (2007) Interneurons of the dentate gyrus: An overview of cell types, terminal fields and neurochemical identity. Prog Brain Res. 163: 217-232. Jacobsen JS, Wu CC, Redwine JM, Comery TA, Arias R, Bowlby M, Martone R, Morrison JH, Pangalos MN, Reinhart PH & Bloom FE (2006) Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A. 103: 5161-5166. Jiang X, West AA & Caudill MA (2014) Maternal choline supplementation: A nutritional approach for improving offspring health? Trends Endocrinol Metab. 25: 263-273. Jinde S, Zsiros V & Nakazawa K (2013) Hilar mossy cell circuitry controlling dentate granule cell excitability. Front Neural Circuits. 7: 14. Kadriu B, Guidotti A, Costa E & Auta J (2009) Imidazenil, a non-sedating anticonvulsant benzodiazepine, is more potent than diazepam in protecting against dfp-induced seizures and neuronal damage. Toxicology. 256: 164-174. Kam K, Duffy AM, Moretto J, LaFrancois JJ & Scharfman HE (2016) Interictal spikes during sleep are an early defect in the Tg2576 mouse model of amyloid neuropathology. Sci Rep. 6: 20119. Kelley CM, Ash JA, Powers BE, Velazquez R, Alldred MJ, Ikonomovic MD, Ginsberg SD, Strupp BJ & Mufson EJ (2016) Effects of maternal choline supplementation on the septohippocampal cholinergic system in the Ts65Dn mouse model of Down syndrome. Curr Alzheimer Res. 13: 84-96. Kelley CM, Ginsberg SD, Alldred MJ, Strupp BJ & Mufson EJ (2019) Maternal choline supplementation alters basal forebrain cholinergic neuron gene expression in the Ts65Dn mouse model of Down syndrome. Dev Neurobiol. 79: 664-683. Kennedy BC, Dimova JG, Siddappa AJ, Tran PV, Gewirtz JC & Georgieff MK (2014) Prenatal choline supplementation ameliorates the long-term neurobehavioral effects of fetal-neonatal iron deficiency in rats. J Nutr. 144: 1858-1865. Kesner RP, Taylor JO, Hoge J & Andy F (2015) Role of the dentate gyrus in mediating object-spatial configuration recognition. Neurobiol Learn Mem. 118: 42-48. Kleen JK, Scott RC, Holmes GL & Lenck-Santini PP (2010) Hippocampal interictal spikes disrupt cognition in rats. Ann Neurol. 67: 250-257. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 Kleen JK, Scott RC, Holmes GL, Roberts DW, Rundle MM, Testorf M, LenckSantini PP & Jobst BC (2013) Hippocampal interictal epileptiform activity disrupts cognition in humans. Neurology. 81: 18-24. Lamoureux JA, Meck WH & Williams CL (2008) Prenatal choline availability alters the context sensitivity of pavlovian conditioning in adult rats. Learn Mem. 15: 866-875. Langley EA, Krykbaeva M, Blusztajn JK & Mellott TJ (2015) High maternal choline consumption during pregnancy and nursing alleviates deficits in social interaction and improves anxiety-like behaviors in the BTBR t+itpr3tf/j mouse model of autism. Behav Brain Res. 278: 210-220. Li X, Chen W, Yu Q, Zhang Q, Zhang T, Huang X, Li H, He A, Yu H, Jing W, Du H, Ke X, Zhang B, Tian Q, Liu R & Lu Y (2021) A circuit of mossy cells controls the efficacy of memory retrieval by gria2i inhibition of gria2. Cell Rep. 34: 108741. Lind D, Franken S, Kappler J, Jankowski J & Schilling K (2005) Characterization of the neuronal marker neun as a multiply phosphorylated antigen with discrete subcellular localization. J Neurosci Res. 79: 295-302. Lisgaras CP & Scharfman HE (2023) Interictal spikes in animal models of alzheimer’s disease: Dominance of the dentate gyrus and cholinergic control by medial septum. Biorxiv. Lobo F, Haase J & Brandhorst S (2022) The effects of dietary interventions on brain aging and neurological diseases. Nutrients. 14. Loy R, Heyer D, Williams CL & Meck WH (1991) Choline-induced spatial memory facilitation correlates with altered distribution and morphology of septal neurons. Adv Exp Med Biol. 295: 373-382. Mao XY, Yin XX, Guan QW, Xia QX, Yang N, Zhou HH, Liu ZQ & Jin WL (2021) Dietary nutrition for neurological disease therapy: Current status and future directions. Pharmacol Ther. 226: 107861. Matsuda S, Umeda M, Uchida H, Kato H & Araki T (2009) Alterations of oxidative stress markers and apoptosis markers in the striatum after transient focal cerebral ischemia in rats. J Neural Transm (Vienna). 116: 395-404. McCall N, Mahadevia D, Corriveau JA & Glenn MJ (2015) Adult emotionality and neural plasticity as a function of adolescent nutrient supplementation in male rats. Pharmacol Biochem Behav. 132: 125-135. McClung CA, Ulery PG, Perrotti LI, Zachariou V, Berton O & Nestler EJ (2004) ∆FosB a molecular switch for long-term adaptation in the brain. Mol Brain Res. 132: 146-154. Meck WH, Smith RA & Williams CL (1988) Pre- and postnatal choline supplementation produces long-term facilitation of spatial memory. Dev Psychobiol. 21: 339-353. Meck WH & Williams CL (1999) Choline supplementation during prenatal development reduces proactive interference in spatial memory. Dev Brain Res. 118: 51-59. Meck WH & Williams CL (2003) Metabolic imprinting of choline by its availability during gestation: Implications for memory and attentional processing across the lifespan. Neurosci Biobehav Rev. 27: 385-399. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 Meck WH, Williams CL, Cermak JM & Blusztajn JK (2007) Developmental periods of choline sensitivity provide an ontogenetic mechanism for regulating memory capacity and age-related dementia. Front Integr Neurosci. 1: 7. Mellott TJ, Williams CL, Meck WH & Blusztajn JK (2004) Prenatal choline supplementation advances hippocampal development and enhances mapk and creb activation. FASEB J. 18: 545-547. Mellott TJ, Huleatt OM, Shade BN, Pender SM, Liu YB, Slack BE & Blusztajn JK (2017) Perinatal choline supplementation reduces amyloidosis and increases choline acetyltransferase expression in the hippocampus of the appsweps1de9 Alzheimer's disease model mice. PLoS One. 12: e0170450. Middei S, Geracitano R, Caprioli A, Mercuri N & Ammassari-Teule M (2004) Preserved fronto-striatal plasticity and enhanced procedural learning in a transgenic mouse model of Alzheimer's disease overexpressing mutant happswe. Learn Mem. 11: 447-452. Moreno HC, de Brugada I, Carias D & Gallo M (2013) Long-lasting effects of prenatal dietary choline availability on object recognition memory ability in adult rats. Nutr Neurosci. 16: 269-274. Morris TA, Jafari N & DeLorenzo RJ (2000) Chronic ∆FosB expression and increased AP-1 transcription factor binding are associated with the long term plasticity changes in epilepsy. Mol Brain Res. 79: 138-149. Mullen RJ, Buck CR & Smith AM (1992) Neun, a neuronal specific nuclear protein in vertebrates. Development. 116: 201-211. Myers CE & Scharfman HE (2009) A role for hilar cells in pattern separation in the dentate gyrus: A computational approach. Hippocampus. 19: 321-337. Myers CE & Scharfman HE (2011) Pattern separation in the dentate gyrus: A role for the CA3 backprojection. Hippocampus. 21: 1190-1215. Olafsson E, Ludvigsson P, Gudmundsson G, Hesdorffer D, Kjartansson O & Hauser WA (2005) Incidence of unprovoked seizures and epilepsy in iceland and assessment of the epilepsy syndrome classification: A prospective study. Lancet Neurol. 4: 627-634. Palop JJ, Chin J, Roberson ED, Wang J, Thwin MT, Bien-Ly N, Yoo J, Ho KO, Yu GQ, Kreitzer A, Finkbeiner S, Noebels JL & Mucke L (2007) Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer's disease. Neuron. 55: 697-711. Palop JJ, Mucke L & Roberson ED (2011) Quantifying biomarkers of cognitive dysfunction and neuronal network hyperexcitability in mouse models of Alzheimer's disease: Depletion of calcium-dependent proteins and inhibitory hippocampal remodeling. Methods Mol Biol. 670: 245-262. Pofahl M, Nikbakht N, Haubrich AN, Nguyen T, Masala N, Distler F, Braganza O, Macke JH, Ewell LA, Golcuk K & Beck H (2021) Synchronous activity patterns in the dentate gyrus during immobility. Elife. 10. Poletaeva, II, Surina NM, Ashapkin VV, Fedotova IB, Merzalov IB, Perepelkina OV & Pavlova GV (2014) Maternal methyl-enriched diet in rat reduced the audiogenic seizure proneness in progeny. Pharmacol Biochem Behav. 127: 21-26. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 Portiansky EL, Barbeito CG, Gimeno EJ, Zuccolilli GO & Goya RG (2006) Loss of neun immunoreactivity in rat spinal cord neurons during aging. Exp Neurol. 202: 519-521. Power R, Prado-Cabrero A, Mulcahy R, Howard A & Nolan JM (2019) The role of nutrition for the aging population: Implications for cognition and Alzheimer's disease. Annu Rev Food Sci Technol. 10: 619-639. Powers BE, Velazquez R, Kelley CM, Ash JA, Strawderman MS, Alldred MJ, Ginsberg SD, Mufson EJ & Strupp BJ (2016) Attentional function and basal forebrain cholinergic neuron morphology during aging in the Ts65Dn mouse model of Down syndrome. Brain Struct Funct. 221: 4337-4352. Powers BE, Kelley CM, Velazquez R, Ash JA, Strawderman MS, Alldred MJ, Ginsberg SD, Mufson EJ & Strupp BJ (2017) Maternal choline supplementation in a mouse model of Down syndrome: Effects on attention and nucleus basalis/substantia innominata neuron morphology in adult offspring. Neuroscience. 340: 501-514. Powers BE, Velazquez R, Strawderman MS, Ginsberg SD, Mufson EJ & Strupp BJ (2021) Maternal choline supplementation as a potential therapy for Down syndrome: Assessment of effects throughout the lifespan. Front Aging Neurosci. 13: 723046. Rausch R, Lieb JP & Crandall PH (1978) Neuropsychologic correlates of depth spike activity in epileptic patients. Arch Neurol. 35: 699-705. Raza SA, Albrecht A, Caliskan G, Muller B, Demiray YE, Ludewig S, Meis S, Faber N, Hartig R, Schraven B, Lessmann V, Schwegler H & Stork O (2017) Hipp neurons in the dentate gyrus mediate the cholinergic modulation of background context memory salience. Nat Commun. 8: 189. Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A & Hen R (2011) Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature. 472: 466-470. Sandstrom NJ, Loy R & Williams CL (2002) Prenatal choline supplementation increases NGF levels in the hippocampus and frontal cortex of young and adult rats. Brain Res. 947: 9-16. Sarkisova KY, Fedosova EA, Shatskova AB, Rudenok MM, Stanishevskaya VA & Slominsky PA (2023) Maternal methyl-enriched diet increases dnmt1, hcn1, and th gene expression and suppresses absence seizures and comorbid depression in offspring of wag/rij rats. Diagnostics (Basel). 13. Sarnat HB, Nochlin D & Born DE (1998) Neuronal nuclear antigen (neun): A marker of neuronal maturation in early human fetal nervous system. Brain Dev. 20: 88-94. Scharfman HE (1999) The role of nonprincipal cells in dentate gyrus excitability and its relevance to animal models of epilepsy and temporal lobe epilepsy. Adv Neurol. 79: 805-820. Scharfman HE, Mercurio TC, Goodman JH, Wilson MA & MacLusky NJ (2003) Hippocampal excitability increases during the estrous cycle in the rat: A potential role for brain-derived neurotrophic factor. J Neurosci. 23: 1164111652. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 Scharfman HE & Myers CE (2012) Hilar mossy cells of the dentate gyrus: A historical perspective. Front Neural Circuits. 6: 106. Scharfman HE (2016) The enigmatic mossy cell of the dentate gyrus. Nat Rev Neurosci. 17: 562-575. Sloviter RS (1994) The functional organization of the hippocampal dentate gyrus and its relevance to the pathogenesis of temporal lobe epilepsy. Ann Neurol. 35: 640-654. Sperk G, Hamilton T & Colmers WF (2007) Neuropeptide Y in the dentate gyrus. Prog Brain Res. 163: 285-297. Spyrka J & Hess G (2018) Repeated neck restraint stress bidirectionally modulates excitatory transmission in the dentate gyrus and performance in a hippocampus-dependent memory task. Neuroscience. 379: 32-44. Strupp BJ, Powers BE, Velazquez R, Ash JA, Kelley CM, Alldred MJ, Strawderman M, Caudill MA, Mufson EJ & Ginsberg SD (2016) Maternal choline supplementation: A potential prenatal treatment for Down syndrome and Alzheimer's disease. Curr Alzheimer Res. 13: 97-106. Vandrey B, Garden DLF, Ambrozova V, McClure C, Nolan MF & Ainge JA (2020) Fan cells in layer 2 of the lateral entorhinal cortex are critical for episodic-like memory. Curr Biol. 30: 169-175 e165. Velazquez R, Ferreira E, Knowles S, Fux C, Rodin A, Winslow W & Oddo S (2019) Lifelong choline supplementation ameliorates Alzheimer's disease pathology and associated cognitive deficits by attenuating microglia activation. Aging Cell. 18: e13037. Velazquez R, Ferreira E, Winslow W, Dave N, Piras IS, Naymik M, Huentelman MJ, Tran A, Caccamo A & Oddo S (2020) Maternal choline supplementation ameliorates Alzheimer's disease pathology by reducing brain homocysteine levels across multiple generations. Mol Psychiatry. 25: 2620-2629. Vogel-Ciernia A & Wood MA (2014) Examining object location and object recognition memory in mice. Curr Protoc Neurosci. 69: 8 31 31-17. Wang Y, Guan X, Chen X, Cai Y, Ma Y, Ma J, Zhang Q, Dai L, Fan X & Bai Y (2019) Choline supplementation ameliorates behavioral deficits and Alzheimer's disease-like pathology in transgenic APP/PS1 mice. Mol Nutr Food Res. 63: e1801407. Wolf A, Bauer B, Abner EL, Ashkenazy-Frolinger T & Hartz AM (2016) A comprehensive behavioral test battery to assess learning and memory in 129s6/Tg2576 mice. PLoS One. 11: e0147733. Wolf HK, Buslei R, Schmidt-Kastner R, Schmidt-Kastner PK, Pietsch T, Wiestler OD & Blumcke I (1996) Neun: A useful neuronal marker for diagnostic histopathology. J Histochem Cytochem. 44: 1167-1171. Won SY, Choi SH & Jin BK (2009) Prothrombin kringle-2-induced oxidative stress contributes to the death of cortical neurons in vivo and in vitro: Role of microglial nadph oxidase. J Neuroimmunol. 214: 83-92. Wong-Goodrich SJ, Glenn MJ, Mellott TJ, Liu YB, Blusztajn JK & Williams CL (2011) Water maze experience and prenatal choline supplementation differentially promote long-term hippocampal recovery from seizures in adulthood. Hippocampus. 21: 584-608. bioRxiv preprint doi: https://doi.org/10.1101/2023.05.12.540428; this version posted May 24, 2023. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 Wu XG, Wang SS, Miao H, Cheng JJ, Zhang SF & Shang YZ (2016) Scutellaria barbata flavonoids alleviate memory deficits and neuronal injuries induced by composited abeta in rats. Behav Brain Funct. 12: 33. Yang Y, Liu Z, Cermak JM, Tandon P, Sarkisian MR, Stafstrom CE, Neill JC, Blusztajn JK & Holmes GL (2000) Protective effects of prenatal choline supplementation on seizure-induced memory impairment. J Neurosci. 20: RC109. Yassine N, Lazaris A, Dorner-Ciossek C, Despres O, Meyer L, Maitre M, Mensah-Nyagan AG, Cassel JC & Mathis C (2013) Detecting spatial memory deficits beyond blindness in Tg2576 Alzheimer mice. Neurobiol Aging. 34: 716-730. You JC, Muralidharan K, Park JW, Petrof I, Pyfer MS, Corbett BF, LaFrancois JJ, Zheng Y, Zhang X, Mohila CA, Yoshor D, Rissman RA, Nestler EJ, Scharfman HE & Chin J (2017) Epigenetic suppression of hippocampal calbindin-d28k by ∆FosB drives seizure-related cognitive deficits. Nat Med. 23: 1377-1383. You JC, Stephens GS, Fu CH, Zhang X, Liu Y & Chin J (2018) Genome-wide profiling reveals functional diversification of ∆FosB gene targets in the hippocampus of an Alzheimer's disease mouse model. PLoS One. 13: e0192508. Zeisel SH & da Costa KA (2009) Choline: An essential nutrient for public health. Nutr Rev. 67: 615-623. Zhang TY, Keown CL, Wen X, Li J, Vousden DA, Anacker C, Bhattacharyya U, Ryan R, Diorio J, O'Toole N, Lerch JP, Mukamel EA & Meaney MJ (2018) Environmental enrichment increases transcriptional and epigenetic differentiation between mouse dorsal and ventral dentate gyrus. Nat Commun. 9: 298.